Merkel cell carcinoma (MCC) is an aggressive neuroendocrine skin cancer with a ∼50% response rate to immune checkpoint blockade (ICB) therapy. To identify predictive biomarkers, we integrated bulk and single-cell RNA sequencing (RNA-seq) with spatial transcriptomics from a cohort of 186 samples from 116 patients, including bulk RNA-seq from 14 matched pairs pre- and post-ICB. In nonresponders, tumors show evidence of increased tumor proliferation, neuronal stem cell markers, and IL1. Responders have increased type I/II interferons and preexisting tissue resident (Trm) CD8 or Vδ1 γδ T cells that functionally converge with overlapping antigen-specific transcriptional programs and clonal expansion of public T-cell receptors. Spatial transcriptomics demonstrated colocalization of T cells with B and dendritic cells, which supply chemokines and costimulation. Lastly, ICB significantly increased clonal expansion or recruitment of Trm and Vδ1 cells in tumors specifically in responders, underscoring their therapeutic importance. These data identify potential clinically actionable biomarkers and therapeutic targets for MCC.

Significance: MCC serves as a model of ICB response. We utilized the largest-to-date, multimodal MCC dataset (n = 116 patients) to uncover unique tumor-intrinsic properties and immune circuits that predict response. We identified CD8 Trm and Vδ1 T cells as clinically actionable mediators of ICB response in major histocompatibility complex–high and –low MCCs, respectively.

Merkel cell carcinoma (MCC) is a highly aggressive primary neuroendocrine skin cancer, with high rates of metastasis and recurrence (1). There are two subtypes of MCC, with different etiologies and phenotypes. In 60% to 80% of MCCs, the Merkel cell polyomavirus (MCPyV) is mutated and clonally integrated (2). They express obligate viral oncoproteins, small and truncated large T antigens, which drive carcinogenesis in part by inactivating p53 and pRB (3). While virus positive (VP) MCCs inactivate pRB and p53 through the expression of the small and large T oncoproteins, virus negative (VN) MCCs achieve the same molecular endpoint through near universal somatic mutation of both RB1 and TP53 (2, 4).

VN-MCCs are immunogenic because of the high number of tumor neoantigens from high mutation burdens. VP-MCCs have a low mutation burden but are immunogenic because they constitutively express viral oncoproteins (4). Shared antigens in VP-MCCs ease identification of tumor-specific T cells across patients (5).

For metastatic disease, immunotherapy is the only Food and Drug Administration–approved therapy that significantly prolongs survival. Unfortunately, many patients have either primary (∼50%) or acquired resistance (up to 25%) to immune checkpoint blockade (ICB; refs. 6, 7). To elucidate potential mechanisms, we compiled the largest MCC cohort with comprehensive multimodal molecular analysis (n = 116 patients), specifically focusing on the tumor microenvironment with bulk RNA sequencing (RNA-seq; n = 71 patients), single-cell RNA-seq (scRNA-seq; n = 360,069 cells from 64 patients), multiplexed immunofluorescence (n = 41 patients), and spatial transcriptomics (n = 20 patients). A total of 46 patients had more than one sample. These methods complement each other, addressing limitations and offering insights across tumor-intrinsic and -extrinsic factors. Our multiomic studies identified clinically actionable biomarkers and therapeutic avenues for MCCs and potentially for other cancers.

Compilation of a Clinically and Molecularly Annotated MCC Cohort

We collected the largest multi-institutional MCC dataset with multimodal molecular characterization of 186 samples across 116 patients including 92 patients with evaluable ICB responses (Table 1; Fig. 1A; Supplementary Tables S1 and S2; “Methods”). These include bulk RNA-seq (n = 85 samples from 71 patients; Supplementary Tables S3–S5; Supplementary Fig. S1A) and a modified form of ECCITE-Seq (n = 54 tumor samples from 48 patients; n = 55 blood samples from 32 patients; Supplementary Tables S6 and S7; Supplementary Fig. S1B), including three samples previously reported (8). Moreover, 44 distinct samples were spatially profiled using multiplex immunofluorescence (mIF) imaging (Akoya Phenoimager), spatial cancer gene or whole transcriptome profiling (Nanostring GeoMx), and single-cell resolution spatial transcriptomics (Nanostring CosMx; Fig. 1B; Supplementary Tables S8–S10; Supplementary Fig. S1C). Overall, 63 patients in our dataset were profiled using two or more modalities (Supplementary Fig. S2).

Table 1.

Clinical characteristics of patients included in this study with odds ratio of how characteristics affect immunotherapy response.

CharacteristicsCRPRTotal respondersSDPDTotal nonrespondersNANo ICBP valueOR95% CI
Gender            
 Male 15 24 39 30 35 10 0.54 1.39 0.49–3.92 
 Female (ref) 10 10 1.00 1.00  
Age            
 ≥65 15 19 34 29 33 12 0.92 0.95 0.38–2.38 
 <65 (ref) 13 11 12 1.00 1.00  
Stage at diagnosis            
 Stage III 13 14 27 22 25 0.99 1.00 0.40–2.54 
 Stage IV 0.75 0.80 0.21–3.00 
 Stage I–II (ref) 14 12 13 1.00 1.00  
Immunocompromised            
 Yes 0.05 0.14 0.02–1.22 
 No (ref) 19 27 46 35 39 14 1.00 1.00  
Viral status            
 Positive 13 13 26 27 30 10 0.33 0.65 0.28–1.52 
 Negative (ref) 14 20 13 15 1.00 1.00  
Type of ICB            
 CTLA-4/PD-1 10 12 0.01 10.76 1.31–88.52 
 PD-L1 16 18 0.02 0.30 0.10–0.87 
 PD-1 (ref) 15 14 29 24 26 1.00 1.00  
Radiation naïve            
 Yes 13 20 20 22 0.55 0.77 0.34–1.76 
 No (ref) 12 15 27 20 23 1.00 1.00  
Chemotherapy naive            
 Yes 15 23 38 31 34 0.55 1.37 0.51–3.69 
 No (ref) 11 1.00 1.00  
ICB naïve            
 Yes 19 25 44 35 38 0.18 2.70 0.65–11.18 
 No (ref) 1.00 1.00  
Known primary            
 Yes 13 25 38 37 40 13 0.30 0.53 0.16–1.72 
 No (ref) 1.00 1.00  
Location of primary            
 Upper extremity 14 10 10 0.58 1.40 0.45–4.38 
 Trunk 0.36 0.4 0.06–2.48 
 Lower extremity 10 13 0.55 0.69 0.22–2.22 
 Unknown 10 0.33 2.00 0.52–7.63 
 Head and neck (ref) 10 12 12 12 1.00 1.00  
CharacteristicsCRPRTotal respondersSDPDTotal nonrespondersNANo ICBP valueOR95% CI
Gender            
 Male 15 24 39 30 35 10 0.54 1.39 0.49–3.92 
 Female (ref) 10 10 1.00 1.00  
Age            
 ≥65 15 19 34 29 33 12 0.92 0.95 0.38–2.38 
 <65 (ref) 13 11 12 1.00 1.00  
Stage at diagnosis            
 Stage III 13 14 27 22 25 0.99 1.00 0.40–2.54 
 Stage IV 0.75 0.80 0.21–3.00 
 Stage I–II (ref) 14 12 13 1.00 1.00  
Immunocompromised            
 Yes 0.05 0.14 0.02–1.22 
 No (ref) 19 27 46 35 39 14 1.00 1.00  
Viral status            
 Positive 13 13 26 27 30 10 0.33 0.65 0.28–1.52 
 Negative (ref) 14 20 13 15 1.00 1.00  
Type of ICB            
 CTLA-4/PD-1 10 12 0.01 10.76 1.31–88.52 
 PD-L1 16 18 0.02 0.30 0.10–0.87 
 PD-1 (ref) 15 14 29 24 26 1.00 1.00  
Radiation naïve            
 Yes 13 20 20 22 0.55 0.77 0.34–1.76 
 No (ref) 12 15 27 20 23 1.00 1.00  
Chemotherapy naive            
 Yes 15 23 38 31 34 0.55 1.37 0.51–3.69 
 No (ref) 11 1.00 1.00  
ICB naïve            
 Yes 19 25 44 35 38 0.18 2.70 0.65–11.18 
 No (ref) 1.00 1.00  
Known primary            
 Yes 13 25 38 37 40 13 0.30 0.53 0.16–1.72 
 No (ref) 1.00 1.00  
Location of primary            
 Upper extremity 14 10 10 0.58 1.40 0.45–4.38 
 Trunk 0.36 0.4 0.06–2.48 
 Lower extremity 10 13 0.55 0.69 0.22–2.22 
 Unknown 10 0.33 2.00 0.52–7.63 
 Head and neck (ref) 10 12 12 12 1.00 1.00  

Abbreviations: CI, confidence interval; CR, complete response; NA, not applicable; OR, odds ratio; PD, progressive disease; PR, partial response; ref, reference; SD, stable disease.

Figure 1.

Differentially expressed genes identify key immune and tumor signatures distinguishing immunotherapy responders and nonresponders. A, Patient sample heatmap of assays and clinical metadata for each sample in our dataset. Each column represents a single patient with the available assays and associated clinical characteristics. In total, 116 patients were included in this study. A total of 101 patients received immunotherapy. Seventy-one patients were assayed by bulk RNA-seq, 64 by scRNA-seq, 41 by mIF, and 20 by spatial transcriptomics (GeoMx and CosMx). VN, Virus-negative; VP, Virus-positive. B, Schematic of the study overview. Patient characteristics, FFPE sample sequencing breakdown with sample counts, single-cell processing workflow with sample counts, and single-cell library breakdown with sample counts. Fifty-four tumor samples and 55 PBMC samples were stained with 45 ADTs and enriched for live, CD45+ cells (tissue), or CD3+ cells (PBMC) via FACS. Cells were processed using 10× Chromium and four sequencing libraries were prepared for each sample: 5′ cDNA, ADT, αβ VDJ, and γδ VDJ. C, Volcano plot of differentially expressed (DE) genes between immunotherapy (IO) responders (R) and nonresponders (NR). Bulk RNA-seq was conducted on 63 FFPE tumor samples from 62 immunotherapy naïve, systemic therapy-naïve patients, all samples prior to evaluable responses to ICB. Differentially expressed genes were identified using the DESeq2 package, with the following covariates: sex, viral status, and tissue. Selected protein-coding genes with a FDR < 0.1 were labeled. A total of 800 genes were upregulated in responders, and 436 genes were upregulated in nonresponders. D, Pathway analysis of DE genes in responders. Only protein-coding genes with FDR < 0.1 were included in the analysis. The top five enriched gene sets from the selected gene set libraries are shown for the responder DE genes. Gene sets were ranked based on the Enrichr combined score, which is a measure of both Fisher’s exact test P value and the z-score of the deviation from the expected rank of the gene set. E, Three gene sets enriched in responders with associated genes are represented on the volcano plot of DE genes from responders and nonresponders. Genes with FDR < 0.1 are labeled. F, Enrichment gene list analysis of DE genes found in nonresponders. Only protein-coding genes with FDR < 0.1 were included in the analysis. The top five enriched gene sets from the selected gene set libraries are shown for nonresponder DE genes. Gene sets were ranked based on the Enrichr combined score. G, Two gene sets enriched in nonresponders with associated genes are represented on the volcano plot of DE genes from responders and nonresponders. Only genes with FDR < 0.1 are labeled.

Figure 1.

Differentially expressed genes identify key immune and tumor signatures distinguishing immunotherapy responders and nonresponders. A, Patient sample heatmap of assays and clinical metadata for each sample in our dataset. Each column represents a single patient with the available assays and associated clinical characteristics. In total, 116 patients were included in this study. A total of 101 patients received immunotherapy. Seventy-one patients were assayed by bulk RNA-seq, 64 by scRNA-seq, 41 by mIF, and 20 by spatial transcriptomics (GeoMx and CosMx). VN, Virus-negative; VP, Virus-positive. B, Schematic of the study overview. Patient characteristics, FFPE sample sequencing breakdown with sample counts, single-cell processing workflow with sample counts, and single-cell library breakdown with sample counts. Fifty-four tumor samples and 55 PBMC samples were stained with 45 ADTs and enriched for live, CD45+ cells (tissue), or CD3+ cells (PBMC) via FACS. Cells were processed using 10× Chromium and four sequencing libraries were prepared for each sample: 5′ cDNA, ADT, αβ VDJ, and γδ VDJ. C, Volcano plot of differentially expressed (DE) genes between immunotherapy (IO) responders (R) and nonresponders (NR). Bulk RNA-seq was conducted on 63 FFPE tumor samples from 62 immunotherapy naïve, systemic therapy-naïve patients, all samples prior to evaluable responses to ICB. Differentially expressed genes were identified using the DESeq2 package, with the following covariates: sex, viral status, and tissue. Selected protein-coding genes with a FDR < 0.1 were labeled. A total of 800 genes were upregulated in responders, and 436 genes were upregulated in nonresponders. D, Pathway analysis of DE genes in responders. Only protein-coding genes with FDR < 0.1 were included in the analysis. The top five enriched gene sets from the selected gene set libraries are shown for the responder DE genes. Gene sets were ranked based on the Enrichr combined score, which is a measure of both Fisher’s exact test P value and the z-score of the deviation from the expected rank of the gene set. E, Three gene sets enriched in responders with associated genes are represented on the volcano plot of DE genes from responders and nonresponders. Genes with FDR < 0.1 are labeled. F, Enrichment gene list analysis of DE genes found in nonresponders. Only protein-coding genes with FDR < 0.1 were included in the analysis. The top five enriched gene sets from the selected gene set libraries are shown for nonresponder DE genes. Gene sets were ranked based on the Enrichr combined score. G, Two gene sets enriched in nonresponders with associated genes are represented on the volcano plot of DE genes from responders and nonresponders. Only genes with FDR < 0.1 are labeled.

Close modal

Bulk RNA-seq Reveals Predictive Biomarkers of Response to ICB

First, we analyzed the bulk RNA-seq of tumors, pre-ICB, from a cohort of 63 patients with 62 patients that were systemic treatment naive to minimize confounding variables related to prior therapy. Within this dataset, there were 31 responders (partial or complete response) and 32 nonresponders (stable and/or progressive disease; Supplementary Fig. S1A; Supplementary Table S3; “Methods”).

Differential gene expression analysis revealed 800 upregulated genes in responders and 436 in nonresponders (P adj < 0.1, DESeq2.0; Fig. 1C; Supplementary Table S11). Many of the genes are characteristic of CD8 T cells [αβ T-cell receptor (TCR), GZMA, CD8A, CD8B], γδ T cells (γδ TCR), B cells (IG genes, CD79A), and plasmacytoid dendritic cells (TLR7, LILRB4). Several molecules were associated with tumor-specific cytotoxic T cells, including markers of T-cell exhaustion (HAVCR2, LAG3, and TIGIT) and cytotoxic activity (GZMB and PRF1). Collectively, these data suggest an enrichment of preexisting CD8 T cells, B cells, and specific myeloid subtypes in responders.

Pathway analysis with Enrichr (9) showed upregulation of IFNγ signaling (Padj. = 2.22e−63), IFNα signaling (Padj. = 8.53e−52), and antigen presentation machinery (Padj. = 2.36e−17), associated with IFNγ signaling in other cancers including melanoma (Fig. 1D and E; Supplementary Table S12; ref. 10).

Nonresponders had upregulated MCC-specific genes, like PAX6 and FOXA1 (Supplementary Table S11). Pathway analysis revealed additional enrichment of genes associated: (i) IL1 processing (11), including IL1A and IL1B, and (ii) E2F targets, which are normally expressed during cell division (Fig. 1F and G; Supplementary Table S12).

scRNA-seq Reveals Cellular Composition of the Tumor Microenvironment and Blood

Next, we conducted scRNA-seq initially on 49 tumor samples derived from 43 patients (Supplementary Table S6; Supplementary Fig. S1B). This included eight samples from four patients across two timepoints (including two pairs of samples collected before and after ICB), four samples from two patients spanning different tissues (skin/lymph node pairs), and 37 single samples. Additionally, we analyzed 55 peripheral blood mononuclear cell (PBMC) samples from 32 patients. This included 21 pre/post-ICB pairs (42 samples), two samples from a third timepoint, and 11 samples that were unmatched. Twenty-six of these samples were matched to patients that also had scRNA-seq on tumor tissue (Fig. 1A and B; Supplementary Figs. S3–S5; Supplementary Table S7). A total of 313,757 cells were recovered from these samples using scRNA-seq. An additional cohort of five post-treatment tumor samples with 46,312 cells were integrated and analyzed separately to investigate the cellular landscape after ICB for a total of 360,069 cells. Following quality control, we identified 27 distinct cell types in the tumor immune microenvironment, including CD8 T cells, CD4 T cells, natural killer (NK) cells, γδ T cells, macrophages, dendritic cells, plasma cells, B cells, and plasmacytoid dendritic cell (pDC) clusters (Fig. 2A and B; Supplementary Figs. S3 and S4A; Supplementary Table S13).

Figure 2.

Response to ICB is associated with tumor cell upregulation of an IFNγ signature and negatively correlated with a tumor cycling signature. A, UMAP projection of 139,845 cells among all 49 tissue samples in single-cell transcriptome datasets with major cell-type annotation. PCA inputs were batched-corrected using the Harmony package and cells were clustered using the standard Seurat pipeline. B, Dot plot showing the scaled expression of the top five marker genes for each cell type annotated in the tissue scRNA-seq dataset. C, Differential gene expression analysis of 60,902 tumor cells pseudobulked by tissue sample between ICB responders and nonresponders was performed using the edgeR LRT test with sex and virus status as a covariate. Thirty-four genes were upregulated in responders, and 25 genes were upregulated in nonresponders. Protein-coding genes with FDR < 0.25 are labeled. D, GSEA of DE genes found in immunotherapy responders (R) and nonresponders (NR). Hallmark gene sets were ranked by normalized enrichment score (NES), with the top five gene sets from responders and nonresponders listed. ***, Padj < 0.001; **, Padj < 0.01; *, Padj < 0.05. E, SCENIC regulon analysis was run on 60,902 tumor cells. The top five regulons differentially expressed in responders (R) and nonresponders (NR) are shown, ranked by FDR, Wilcoxon test. F, Scatter plot of CD45+ infiltration (% CD45+ cells from FACS) with logit transformation vs. Hallmark interferon gamma signature module score in tumor cells from scRNA-seq, which depicts a positive Pearson correlation between IFNγ response in the tumor and overall immune infiltration. Samples are colored according to ICB response. IFNG score was calculated on tumor cells by the ModuleScore function in Seurat using the MSIGDB “Hallmark interferon gamma response” genes as input. G, A violin plot of IFNG expression of each cell type split by samples and ranked by mean expression. H, Scatter plots of immune proportion (% CD45+ cells from FACS) with logit transformation or tumor cell IFNG score compared to the proportion of proliferating tumor cells among the total cells with log transformation in each scRNA-seq tissue sample. Samples are colored by ICB response, which shows a negative Pearson correlation between the proportion of proliferating tumor cells and either the immune infiltration or IFNG response in the tumor cells, with nonresponders clustered in the high proliferation, low IFNG or low CD45+ % quartile. I, A Kaplan–Meier curve of post-ICB survival shows that samples with higher levels of tumor cycling cell signature (derived from scRNA-seq) had lower survival. Samples from the bulk RNA-seq dataset were scored based on the “tumor cycling” cell type signature derived from our single cell data with gene set variance analysis (GSVA) and split into the top or bottom quartile (see “Methods”). Statistical test: Cox proportional hazards.

Figure 2.

Response to ICB is associated with tumor cell upregulation of an IFNγ signature and negatively correlated with a tumor cycling signature. A, UMAP projection of 139,845 cells among all 49 tissue samples in single-cell transcriptome datasets with major cell-type annotation. PCA inputs were batched-corrected using the Harmony package and cells were clustered using the standard Seurat pipeline. B, Dot plot showing the scaled expression of the top five marker genes for each cell type annotated in the tissue scRNA-seq dataset. C, Differential gene expression analysis of 60,902 tumor cells pseudobulked by tissue sample between ICB responders and nonresponders was performed using the edgeR LRT test with sex and virus status as a covariate. Thirty-four genes were upregulated in responders, and 25 genes were upregulated in nonresponders. Protein-coding genes with FDR < 0.25 are labeled. D, GSEA of DE genes found in immunotherapy responders (R) and nonresponders (NR). Hallmark gene sets were ranked by normalized enrichment score (NES), with the top five gene sets from responders and nonresponders listed. ***, Padj < 0.001; **, Padj < 0.01; *, Padj < 0.05. E, SCENIC regulon analysis was run on 60,902 tumor cells. The top five regulons differentially expressed in responders (R) and nonresponders (NR) are shown, ranked by FDR, Wilcoxon test. F, Scatter plot of CD45+ infiltration (% CD45+ cells from FACS) with logit transformation vs. Hallmark interferon gamma signature module score in tumor cells from scRNA-seq, which depicts a positive Pearson correlation between IFNγ response in the tumor and overall immune infiltration. Samples are colored according to ICB response. IFNG score was calculated on tumor cells by the ModuleScore function in Seurat using the MSIGDB “Hallmark interferon gamma response” genes as input. G, A violin plot of IFNG expression of each cell type split by samples and ranked by mean expression. H, Scatter plots of immune proportion (% CD45+ cells from FACS) with logit transformation or tumor cell IFNG score compared to the proportion of proliferating tumor cells among the total cells with log transformation in each scRNA-seq tissue sample. Samples are colored by ICB response, which shows a negative Pearson correlation between the proportion of proliferating tumor cells and either the immune infiltration or IFNG response in the tumor cells, with nonresponders clustered in the high proliferation, low IFNG or low CD45+ % quartile. I, A Kaplan–Meier curve of post-ICB survival shows that samples with higher levels of tumor cycling cell signature (derived from scRNA-seq) had lower survival. Samples from the bulk RNA-seq dataset were scored based on the “tumor cycling” cell type signature derived from our single cell data with gene set variance analysis (GSVA) and split into the top or bottom quartile (see “Methods”). Statistical test: Cox proportional hazards.

Close modal

Types I and II Interferon Signatures in Tumor Cells Predict Response

First, we analyzed the 60,902 tumor cells, which were marked by the expression of neuroendocrine transcription factor ATOH1 and chromogranin (3). We examined expression of the viral oncogenes small T antigen (sTAg) or large T antigen (LTAg) by scRNA-seq and bulk RNA-seq. Twenty six of 43 patients had samples with multiple reads for both sTAg and LTAg and were thus considered VP. The remainder had no transcriptional evidence of MCPyV sTAg or LTAg expression, which we labeled VN.

Next, we performed differential gene and pathway expression analyses (Supplementary Table S14; Fig. 2C). In responders, we observed upregulation of multiple pathways. Similar to bulk RNA-seq, there was upregulation of genes associated with IFNγ (STAT1, MX1, IRF1) as well as IFNα (OAS1, IFI27, IRF9). Again, many IFNγ-associated genes are implicated in antigen presentation (i.e., HLA-I, HLA-II, ERAP2; Fig. 2D; Supplementary Table S15). Next, we ran SCENIC regulon analysis to identify significant transcription factor–associated transcriptional networks. Top regulons in responders highlighted type I/II interferon responses (IRF7, IRF9, STAT1, IRF1; Fig. 2E; Supplementary Table S16).

We calculated a module score to quantify IFNγ and IFN responses using previously described methods (10, 12). IFNα and IFNγ response module scores were highly correlated (Pearson correlation, R2 = 0.928; P < 1e−10). Both signatures were associated with the infiltration of immune cells, suggestive of preexisting immunity (Fig. 2F). The source of type I interferons was not clear because it was not well captured by scRNA-seq. However, subsequent analyses showed that the IFNα module score in multiple cell types (tumor cells, T/NK cells, and stroma) was correlated with the presence of pDCs, which express high levels of type I interferons (Pearson correlation, R = 0.38; P = 0.011 in tumor cells; Supplementary Fig. S6A and S6B; ref. 13). Other possible candidate cell types showed lower correlations (classical dendritic cell (cDC) R = 0.045; macrophage R = −0.0010). The source of IFNγ appeared to be primarily CD8 and other NK/T-cells (Fig. 2G; Supplementary Fig. S7A). The enrichment of type I/II interferon gene sets in responders did not depend on the site of tissue sampling (LN vs. skin) or metastatic status (primary vs. metastasis; Supplementary Fig. S7B).

Tumor Cell Proliferation Is Anticorrelated with Immune Cell Infiltration and Survival Post ICB

In nonresponders, we found upregulation of 21 genes and eight Hallmark pathways via Gene Set Enrichment Analysis (GSEA). These include MYC (Padj = 3.05e−17), mTORC1 (Padj = 1.68e−10), and E2F (Padj = 7.62e−4). We hypothesized that the E2F signatures observed in bulk RNA-seq arose from tumor cells. To test this hypothesis, we first identified cycling tumor cells. These cells expressed markers of cell cycle/division, MKI67, TOP2A, CENPF, and E2F targets, such as CCNB2 and CDKN3, varying in proportion across samples from 0% to 5% of the total tumor cells. Because mTORC1 and MYC can be associated with proliferation, we confirmed that they retained significance even when controlled for cell cycle (Supplementary Fig. S8A and S8B; “Methods”).

Consistent with data from other tumor types (14), we observed that proliferating tumor cells was negatively correlated with both overall immune infiltration (Pearson correlation, R = −0.6; P = 1e−5) and the IFNγ response, which we posit is a response to IFNγ producing lymphocytes in the TME (Pearson correlation, R = −0.6, P = 4e−5; Fig. 2H). Finally, to test the effects on survival, we derived a tumor cycling signature and applied it to our bulk RNA-seq, which is significantly larger than our scRNA-seq dataset. Samples from the bulk RNA-seq dataset were scored based on the “tumor cycling” cell type signature derived from our single cell dataset with gene set variance analysis (GSVA) and split into the top or bottom quartile (see “Methods”). Patients with high levels of the tumor cycling signature had worse overall survival post ICB (Fig. 2I; Supplementary Table S17; P = 0.016, Cox proportional hazard).

To find additional drivers of primary resistance, we analyzed transcription factors upregulated in tumors from nonresponders. The most significantly upregulated gene in nonresponders was OTX2, a transcription factor involved in neural cell development. SCENIC analysis (15) confirmed that OTX2 target genes are also upregulated (Fig. 2E). The analysis identified additional transcription factors associated with neural stem cells (HOXA10, GATA2, and SIX1; Supplementary Table S16; refs. 1618).

Collectively, tumor cell analysis identified genes and signatures predictive of the response to ICB that correlate with the numbers of immune cells and effector molecules produced by them in the tumor microenvironment. To assess this further, we analyzed the immune infiltrates.

CD8 T-cell Analysis Reveals Two Independent Trajectories

We turned next to CD8 T cells because they express the highest levels of IFNγ in the tumor microenvironment (Fig. 2G; Supplementary Fig. S7B). Single-cell analysis identified 15 distinct clusters that were organized into two distinct metaclusters (Fig. 3A). The first, which we termed circulating T cells (Tcirc), included CD8 clusters C00_CD8, C01_CD8, C02_CD8, C09_CD8, C11_CD8, and C14_CD8. This cluster is enriched in T cells normally found in circulation, e.g., naïve, central memory, and terminally differentiated effector memory (Temra) T cells. Cells within this metacluster have upregulated genes or proteins associated with naïve and central memory cells. This includes markers of stemness, e.g., IL7R, TCF7, and LTB (19) markers of blood circulation, e.g., CCR7, SELL, S1PR1, and KLF2 (19). Changes in a subset of genes was confirmed at the protein level including CD127, CD62L, and CD45RA, which are upregulated in naïve, central memory, and Temra cells (Supplementary Table S18; Fig. 3B–D).

Figure 3.

CD8 TILs form two distinct developmental trajectories composed of circulatory T cells and Tissue resident memory subsets. A, UMAP projections of 24,448 CD8 T cells in 15 cell clusters. Outlines of the clusters show the formation of two metaclusters: Tcirc and Trm cells. Cells were a subset from all tissue scRNA-seq samples and clustered using the standard Seurat pipeline. B, Heatmap of the ADTs protein average scaled expression of each CD8 cluster with hierarchal clustering into two major metaclusters distinguishing Tcirc and Trm cells. C, Bar chart of the proportion of co-expressing, co-inhibitory markers statistically enriched on Trm compared to Tcirc cells (χ2, P < 2.2e−16). D, Dot plot of selected average marker gene expression for each CD8 lineage with functionally grouped genes. logFC: log2 fold change. E, Diffusion mapping projection of CD8 T cells with cell cycle genes regressed out using the Destiny package. The diffusion map shows two separate lineages, which are colored by Trm or Tcirc. Cells in the CD8 cluster C04 are depicted in gray. C04 show characteristics of both Trm and Tcirc cells. DC, diffusion component. F, Heatmap of the log-likelihood ratio (LLR) of shared TCR clones between CD8 clusters with the heatmap clustered by LLR demonstrates the formation of two metaclusters that match the Trm and Tcirc lineages. *, P < 0.05, Fisher’s test. G, Bar chart of all expanded CD8 clonotypes in the tissue dataset. Each line represents the proportion of cells in each lineage of the single clonotype. The clonotypes were ranked from the highest proportion of cells in the Trm lineage to the lowest proportion (highest in the Tcirc lineage). For 88% of the clonotypes, 100% of the cells were restricted to either the Trm or Tcirc lineage. H, Bar chart of the proportion of clones that were also found in the PBMC single-cell dataset split by developmental lineage. Only matched blood samples were included in the analysis. χ2 test for significant differences between proportions. I, UMAP projection (same as A) colored by the TCR clone size. J, Violin plots of clonal size of all clonotypes found in the Trm or Tcirc lineage.

Figure 3.

CD8 TILs form two distinct developmental trajectories composed of circulatory T cells and Tissue resident memory subsets. A, UMAP projections of 24,448 CD8 T cells in 15 cell clusters. Outlines of the clusters show the formation of two metaclusters: Tcirc and Trm cells. Cells were a subset from all tissue scRNA-seq samples and clustered using the standard Seurat pipeline. B, Heatmap of the ADTs protein average scaled expression of each CD8 cluster with hierarchal clustering into two major metaclusters distinguishing Tcirc and Trm cells. C, Bar chart of the proportion of co-expressing, co-inhibitory markers statistically enriched on Trm compared to Tcirc cells (χ2, P < 2.2e−16). D, Dot plot of selected average marker gene expression for each CD8 lineage with functionally grouped genes. logFC: log2 fold change. E, Diffusion mapping projection of CD8 T cells with cell cycle genes regressed out using the Destiny package. The diffusion map shows two separate lineages, which are colored by Trm or Tcirc. Cells in the CD8 cluster C04 are depicted in gray. C04 show characteristics of both Trm and Tcirc cells. DC, diffusion component. F, Heatmap of the log-likelihood ratio (LLR) of shared TCR clones between CD8 clusters with the heatmap clustered by LLR demonstrates the formation of two metaclusters that match the Trm and Tcirc lineages. *, P < 0.05, Fisher’s test. G, Bar chart of all expanded CD8 clonotypes in the tissue dataset. Each line represents the proportion of cells in each lineage of the single clonotype. The clonotypes were ranked from the highest proportion of cells in the Trm lineage to the lowest proportion (highest in the Tcirc lineage). For 88% of the clonotypes, 100% of the cells were restricted to either the Trm or Tcirc lineage. H, Bar chart of the proportion of clones that were also found in the PBMC single-cell dataset split by developmental lineage. Only matched blood samples were included in the analysis. χ2 test for significant differences between proportions. I, UMAP projection (same as A) colored by the TCR clone size. J, Violin plots of clonal size of all clonotypes found in the Trm or Tcirc lineage.

Close modal

The second metacluster (comprising CD8 clusters C03_CD8, C04_CD8, C05_CD8, C06_CD8, C07_CD8, C08_CD8, C10_CD8, C12_CD8, and C13_CD8) showed the upregulation of genes associated with tissue-resident T (Trm) differentiation. These include adhesion genes (ITGAE, ITGA1, CD69), chemokine receptors normally found in skin-resident T cells (CXCR6), transcription factors (PRDM1 and RUNX3; ref.20), and skin Trm-specific metabolic genes (FABP5; Fig. 3D; Supplementary Table S19; ref. 21).

Trm cells displayed significantly increased expression of genes reflecting recent activation (41BB, CD38), as well as markers of chronic activation/exhaustion (LAG3, HAVCR2, PDCD1, ENTPD1, TOX; Fig. 3C; χ2; P < 2.2e−16). Based on the antibody-derived tags (ADT), the most common surface phenotype of Trm cells in MCCs was CD103+, CD137+, CD39+, CD279+, TIGIT+, CD8 T cells (Fig. 3B). With the addition of RNA markers, the majority of Trm cells also expressed LAG3 and HAVCR2 (Fig. 3C and D). Trm cells had over 2-fold increased expression of IFNγ compared to Tcirc cells, with clusters C03_CD8, C05_CD8, C07_CD8, and C13_CD8 showing the highest levels (Supplementary Table S20; Supplementary Fig. S9). They also showed the highest expression of cytotoxic enzymes including GZMB and PRF1 (Fig. 3B–D).

To elucidate the relationship between Trm and Tcirc metaclusters, we performed two analyses. First, diffusion mapping revealed two distinct developmental lineages (Fig. 3E), which largely mirrored Trm and Tcirc metaclusters. Second, we performed clonotype linkage analysis, which identifies CD8 clusters that have shared TCR clones and are thus developmentally linked. Hierarchical clustering highlighted the two Trm and Tcirc metaclusters (Supplementary Table S21; Fig. 3F). Analysis of individual TCRs underscored the lack of inter-mixing, as 88% of expanded T-cell clones were exclusively restricted to either the Trm or Tcirc metacluster (Fig. 3G), which is unlikely to occur by chance alone (Monte-Carlo simulation, P < 1e6; Supplementary Fig. S10A and S10B).

To confirm that Tcirc cells were derived from the blood, we compared the clonotypic overlap in patient-matched scRNA-seq from the blood. Consistent with our expectation, we found that Tcirc cells shared 4.95-fold more clonotypes with CD8 T cells found in the blood than Trm cells (P < 2.2e−16, χ2; Fig. 3H).

Pseudotime analysis of tissue CD8 T cells provided further insights into the molecular characteristics of these trajectories. We found that Tcirc cells differentiated from a naïve state to a circulating effector memory state, exhibiting decreased expression of naïve markers IL7R and CCR7, increased expression of GZMH, and a subsequent spike in GZMK expression, coinciding with JUND expression (Supplementary Fig. S11A and S11B). Trm cells acquired exhaustion markers during differentiation (TOX, PDCD1) and lost the expression of genes such as BTG1, which may function as a negative regulator of effector activity (Supplementary Fig. S11C and S11D; ref. 22).

Trm Cells Are Tumor-Specific and are Prognostic of ICB Response

We hypothesized that Trm CD8 T cells would be enriched for tumor antigen-specific cells because they showed the highest expression of both activation/exhaustion markers (proteins 41BB, CD38, PD-1, and the TOX gene) and cytotoxic enzymes (GZMA and GZMB; Fig. 3B–D). Moreover, the Trm lineage showed significantly higher levels of clonal expansion (Supplementary Table S22; Fig. 3I and J; Supplementary Fig. S12A and S12B).

To test this, we measured the NeoTCR8 score, a gene signature previously found to be enriched in tumor neoantigen-reactive T cells in multiple solid tumor types (23). The highest NeoTCR8 scores were found in the Trm clusters, with all but one of the Trm clusters ranked higher than any of the Tcirc clusters (Fig. 4A; Supplementary Fig. S9). Importantly, we found that this signature was similar across VP and VN-MCCs and was independent of the site of tumor sampling or stage (Supplementary Fig. S12C and S12D; Supplementary Fig. S13A–S13C).

Figure 4.

CD8 Trm cells are tumor specific and prognostic of ICB response. A, Violin plot of cellular expression of the NeoTCR8 (23) module score split by CD8 cluster. Clusters are colored by developmental lineage: Tcirc or Trm. B, Cell density plot showing the distribution of cells with the MO_MCC_095c1 TCR clonotype, a highly expanded Trm TCR clone found in virus-positive MCC samples. Outline shows the distribution of Tcirc and Trm cells. C, Jurkat-76 cells modified with an NFAT-GFP reporter and CD8α/CD8β were transduced and selected to express the MO_MCC_095c1 TCR. These cells were co-cultured with an autologous lymphoblastoid cell line loaded with pooled peptides from the small and large T antigens of Merkel cell polyoma virus. After co-culture, GFP expression on the reporter cells was measured as a measure of antigen recognition, leading to T-cell activation. CEF (CMV, EBV, Influenza) peptide pool, pool 1, and pool 2, pool 3 are representative examples of negative hits, whereas pools 8 and 12 are positive hits. See Supplementary Fig. S14 shows the complete results for all pools. *, P < 0.05, One-way ANOVA. D, Structural model of the MO_MCC_095c1 TCR in complex with its target peptide, small T (sT) antigen residues 31–39, bound to its putative MHC allele, HLA B*07:02, reveals a canonical TCR-pMHC binding mode and multiple polar contacts that stabilize the interaction (dashed lines). TCRα is magenta, TCRβ cyan, peptide slate (stick representation), and MHC green. The high confidence of the model by AlphaFold score (0.82 out of 1) indicates a high likelihood of structural accuracy. E, Scatter plot of all significant GLIPH2 (26) complementary-determining region 3 (CDR3) motifs found in the CD8 tissue TCR dataset. Axis has been adjusted to show motifs that are >80% found in virus-positive samples and >80% found in Trm cells. These virus-positive (VP) Trm motifs are shared between multiple patients and are highly expanded and represent likely shared anti-MCC TCR motifs. “%” indicates a variable amino acid found in the GLIPH motif. F, Representative mIF images show skin resident CD8 T cells (CD3+CD8+CD103+) infiltrating the tumor parenchyma, marked by pan-cytokeratin (PanCK), and are cytotoxic, marked by granzyme B (GZMB). Scale bar, 100 µm. G, A scatter plot of tissue-resident CD8 T-cell densities (percentage of CD3+CD8+ expressing CD103 per mm2) from 29 mIF samples from 26 patients (17 responders, 9 nonresponders). Wilcoxon test. H, A violin plot of the ratio of Trm to Tcirc cells split by response to immunotherapy (IO). Wilcoxon test. I, Kaplan–Meier curve of survival from the start of ICB from the bulk RNA-seq dataset. Samples were split based on the top and bottom quartiles of the Trm gene set variation analysis score derived from our single-cell dataset (see “Methods” section). Statistical test: Cox proportional hazards.

Figure 4.

CD8 Trm cells are tumor specific and prognostic of ICB response. A, Violin plot of cellular expression of the NeoTCR8 (23) module score split by CD8 cluster. Clusters are colored by developmental lineage: Tcirc or Trm. B, Cell density plot showing the distribution of cells with the MO_MCC_095c1 TCR clonotype, a highly expanded Trm TCR clone found in virus-positive MCC samples. Outline shows the distribution of Tcirc and Trm cells. C, Jurkat-76 cells modified with an NFAT-GFP reporter and CD8α/CD8β were transduced and selected to express the MO_MCC_095c1 TCR. These cells were co-cultured with an autologous lymphoblastoid cell line loaded with pooled peptides from the small and large T antigens of Merkel cell polyoma virus. After co-culture, GFP expression on the reporter cells was measured as a measure of antigen recognition, leading to T-cell activation. CEF (CMV, EBV, Influenza) peptide pool, pool 1, and pool 2, pool 3 are representative examples of negative hits, whereas pools 8 and 12 are positive hits. See Supplementary Fig. S14 shows the complete results for all pools. *, P < 0.05, One-way ANOVA. D, Structural model of the MO_MCC_095c1 TCR in complex with its target peptide, small T (sT) antigen residues 31–39, bound to its putative MHC allele, HLA B*07:02, reveals a canonical TCR-pMHC binding mode and multiple polar contacts that stabilize the interaction (dashed lines). TCRα is magenta, TCRβ cyan, peptide slate (stick representation), and MHC green. The high confidence of the model by AlphaFold score (0.82 out of 1) indicates a high likelihood of structural accuracy. E, Scatter plot of all significant GLIPH2 (26) complementary-determining region 3 (CDR3) motifs found in the CD8 tissue TCR dataset. Axis has been adjusted to show motifs that are >80% found in virus-positive samples and >80% found in Trm cells. These virus-positive (VP) Trm motifs are shared between multiple patients and are highly expanded and represent likely shared anti-MCC TCR motifs. “%” indicates a variable amino acid found in the GLIPH motif. F, Representative mIF images show skin resident CD8 T cells (CD3+CD8+CD103+) infiltrating the tumor parenchyma, marked by pan-cytokeratin (PanCK), and are cytotoxic, marked by granzyme B (GZMB). Scale bar, 100 µm. G, A scatter plot of tissue-resident CD8 T-cell densities (percentage of CD3+CD8+ expressing CD103 per mm2) from 29 mIF samples from 26 patients (17 responders, 9 nonresponders). Wilcoxon test. H, A violin plot of the ratio of Trm to Tcirc cells split by response to immunotherapy (IO). Wilcoxon test. I, Kaplan–Meier curve of survival from the start of ICB from the bulk RNA-seq dataset. Samples were split based on the top and bottom quartiles of the Trm gene set variation analysis score derived from our single-cell dataset (see “Methods” section). Statistical test: Cox proportional hazards.

Close modal

VP MCCs showed larger TCR clone sizes among CD8 T cells compared to VN MCC. This may be because of a restricted tumor antigen space due to uniform expression of immunogenic viral proteins and few, if any, private somatic mutations/neoantigens (4). This feature facilitated functional validation. We transduced Jurkat-76 cells (αβ TCR knockout) with an NFAT-GFP reporter with four highly expanded TCRs (three Trm-restricted and one Tcirc-restricted) and co-cultured them with autologous lymphoblastoid cell lines (LCL) pulsed with peptide pools. These pools contain 116-amino acid peptides that tile along both the small T antigen and the large T antigen of MCPyV (Supplementary Table S23; ref. 24). All Trm-restricted TCRs demonstrated antigen-specific activation in response to a unique peptide antigen, confirming viral specificity (Fig. 4B and C; Supplementary Fig. S14A–S14D). The expanded Tcirc TCR did not show any specific activation (Supplementary Fig. S14E). Structural modeling supported the antigen specificities of the TCRs tested. When the TCR sequences were modeled with the experimentally validated peptides and the associated predicted MHC, 2/3 Trm TCRs had a TCRmodel2 score >0.8 out of 1, indicating high confidence of binding of the TCR to the functionally validated peptide-MHC complex (Fig. 4D; Supplementary Fig. S14F; ref. 25).

To link TCRs to public Merkel cell polyomavirus antigens within and across patients with VP-MCCs, we deployed GLIPH2, a computational pipeline that identifies biochemical similarities across complementarity-determining regions 3 (CDR3; ref. 26). We found 10 GLIPH2 motifs among 23 unique TCRs that were highly restricted to the VP cohort (>85%) and were highly expanded (>20 clones), suggesting that they respond to tumor-specific Merkel cell polyomavirus antigens (Fig. 4E). These TCRs were also highly enriched in the Trm cells (Supplementary Table S24). In contrast, we found fewer GLIPH motifs (n = 5) enriched in the VN cohort. Unlike for VP MCC, none of these motifs was expanded (i.e., clones of more than one cell) in multiple patients (Supplementary Table S24). This is consistent with the relative absence of public antigens in VN-MCCs where private mutations are created by UV-mediated mutagenesis.

To further explore the antigen specificity of MCC tumor-infiltrating lymphocytes (TIL), we compared our TCR sequences with known TCRs epitopes from three TCR databases that have compiled TCRs with known specificity to common viral pathogens that are not associated with MCC (2729). We found 128 clonotypes that were predicted to bind to public viral antigens not expressed by Merkel cell carcinomas (including cytomegalovirus, influenza, and SARS-CoV-2 but not MCPyV). Consistent with the absence of these antigens in MCC tumor cells, these TCRs showed minimal expansion in the tumor microenvironment (Supplementary Table S25). When present, these clonotypes were significantly enriched in the Tcirc cell trajectory (χ2 = 19.4; P = 0.00001; Supplementary Fig. S15A).

Finally, we assessed the clinical significance of Trm cells. First, the prevalence of Trm cells was correlated with response to ICB (P = 0.018; Supplementary Fig. S15B). This was corroborated by multiplex immunofluorescence (mIF) data performed on 44 samples from 38 patients (24 with assessable response), of which 17 had associated tumor scRNA-seq. No one marker set defines Trm cells; however, our single-cell data suggest that CD103 is expressed in 38.6%, a plurality of Trm cells. CD3+CD8+CD103+ skin-resident T cells were observed infiltrating the tumor parenchyma (Fig. 4F), and tumors from responders had significantly higher densities of CD3+CD8+CD103+ cells as compared to nonresponders in our mIF panel (Fig. 4G).

Strikingly, the ratio of Trm to Tcirc cells was also predictive of the clinical outcomes. The ratio of Trm:Tcirc was higher in responders, with all nonresponders having equal or more Tcirc cells compared to Trm cells. (Fig. 4H). This was confirmed in the bulk RNA-seq using GSVA on a Trm signature we derived from our single-cell dataset (see “Methods”). Patients with tumors with high expression of the Trm signature (the top quartile) had significantly improved median survival from the start of ICB compared to those with low Trm signatures (the bottom quartile; median survival not reached vs. 578 days, respectively, P = 0.038, Cox proportional hazard; Fig. 4I). Collectively, these data show that the presence of Trm cells is associated with both response to ICB and longer survival.

Vδ1 γδ T Cells Are Enriched in Tumors of Responders with Low IFNγ Signatures

Next, we focused on samples from patients who responded to immunotherapy despite having a low IFNγ signature (Fig. 5A). This analysis identified non-conventional cytotoxic T cells as potential prognostic biomarkers. Single-cell analysis identified 17 clusters of γδ and NK cells (Fig. 5B). To unequivocally identify γδ T cells, we utilized TCR data obtained through our modification of ECCITE-seq (30), which enriches γδ TCR sequences. With one exception, most patients had low levels of NK cells (<2% of cells). In contrast, we found that γδ T cells were enriched (5.7%–25.0% of all cells) in 7/49 of tumor samples, including four samples from responders with a low IFNγ signature (Fig. 5A). We observed that tumors with low MHC-I expression were enriched for expression of TRDV1, which encodes Vδ1 TCR (Fig. 5C).

Figure 5.

Landscape of γδ T cells and NK cells in MCC mirrors Trm and Tcirc trajectories found in CD8 T cells suggesting functional convergence in T-cell driven responses. A, Scatter plot of the interferon-γ response score (module score from the hallmark gene set) of tumor cells vs. the proportion of γδ T cells in a sample that shows the subset of γδ T cell-enriched samples that also have a low IFNG score. B, UMAP projection of 9722 γδ/NK cells among 16 clusters from tissue scRNA-seq data. Cells were clustered using the standard Seurat pipeline. C, Differentially expressed genes (pseudobulked, edgeR LRT test) in γδ/NK cells compared to samples with low tumor HLA I expression to samples with high tumor HLA I expression. D, A chord diagram of γ and δ V chain pairings for all cells in the tissue scRNA-seq dataset. E, Dot plot of average expression of selected significant, Wilcoxon gene markers of γδ and NK cells in both the tissue datasets separated by developmentally distinct δ-chain or NK lineage. δ1 and δ3 T cells show higher levels of exhaustion markers, activation markers, KIR family members, cytotoxic effectors, CXCR6, adhesion molecules, and exhaustion-related transcription factors (TF). δ2 T cells express high levels of TNF, LTB, naïve/T central memory (Tcm) markers, and AP-1 family members (FOS). F, Violin plot of selected significant Wilcoxon protein markers separated by tissue/ PBMC samples and grouped by δ-chain or NK lineage. G, A volcano plot of differentially expressed protein markers (ADT) of Vδ1 cells vs. Vδ2 cells. Vδ1 cells upregulated exhaustion and activation markers. Vδ2 cells upregulated naïve and T central memory markers. All labeled proteins had Padj. < 0.05. Wilcoxon test. H, Multiple alignment and consensus sequence of δ CDR3 of the top expanded Vδ1 clones shows clonal focusing with sequence similarity between patients. See “Methods” for details on TCR selection. I, UMAP projection of Vδ1 cells (same projection as B) colored by pseudotime as calculated by Monocle demonstrate that “root” cells are found mainly in C08_Vd1 whereas differentiated cells are found in C02_Vd1. Root cells were selected based on their highest expression of naïve-like markers, including TCF7, CD45RA, CD127, and CCR7. J, Heatmap of genes that are significantly and nonlinearly correlated with Vδ1 pseudotime. Vδ1 T cells have high expression of naïve markers like TCF7 as well as AP-1 family members at the beginning of pseudotime. During differentiation, Vδ1 cells upregulate co-inhibitory, cytotoxic, activating, and Trm makers. Genes are colored by scaled expression across pseudotime. K, Representative image of mIF showing cytotoxic granzyme B (GZMB+) γδ T cells δ TCR (dTCR+) infiltrating the tumor parenchyma, marked by PanCK, as indicated by arrows. Scale bar, 50 microns.

Figure 5.

Landscape of γδ T cells and NK cells in MCC mirrors Trm and Tcirc trajectories found in CD8 T cells suggesting functional convergence in T-cell driven responses. A, Scatter plot of the interferon-γ response score (module score from the hallmark gene set) of tumor cells vs. the proportion of γδ T cells in a sample that shows the subset of γδ T cell-enriched samples that also have a low IFNG score. B, UMAP projection of 9722 γδ/NK cells among 16 clusters from tissue scRNA-seq data. Cells were clustered using the standard Seurat pipeline. C, Differentially expressed genes (pseudobulked, edgeR LRT test) in γδ/NK cells compared to samples with low tumor HLA I expression to samples with high tumor HLA I expression. D, A chord diagram of γ and δ V chain pairings for all cells in the tissue scRNA-seq dataset. E, Dot plot of average expression of selected significant, Wilcoxon gene markers of γδ and NK cells in both the tissue datasets separated by developmentally distinct δ-chain or NK lineage. δ1 and δ3 T cells show higher levels of exhaustion markers, activation markers, KIR family members, cytotoxic effectors, CXCR6, adhesion molecules, and exhaustion-related transcription factors (TF). δ2 T cells express high levels of TNF, LTB, naïve/T central memory (Tcm) markers, and AP-1 family members (FOS). F, Violin plot of selected significant Wilcoxon protein markers separated by tissue/ PBMC samples and grouped by δ-chain or NK lineage. G, A volcano plot of differentially expressed protein markers (ADT) of Vδ1 cells vs. Vδ2 cells. Vδ1 cells upregulated exhaustion and activation markers. Vδ2 cells upregulated naïve and T central memory markers. All labeled proteins had Padj. < 0.05. Wilcoxon test. H, Multiple alignment and consensus sequence of δ CDR3 of the top expanded Vδ1 clones shows clonal focusing with sequence similarity between patients. See “Methods” for details on TCR selection. I, UMAP projection of Vδ1 cells (same projection as B) colored by pseudotime as calculated by Monocle demonstrate that “root” cells are found mainly in C08_Vd1 whereas differentiated cells are found in C02_Vd1. Root cells were selected based on their highest expression of naïve-like markers, including TCF7, CD45RA, CD127, and CCR7. J, Heatmap of genes that are significantly and nonlinearly correlated with Vδ1 pseudotime. Vδ1 T cells have high expression of naïve markers like TCF7 as well as AP-1 family members at the beginning of pseudotime. During differentiation, Vδ1 cells upregulate co-inhibitory, cytotoxic, activating, and Trm makers. Genes are colored by scaled expression across pseudotime. K, Representative image of mIF showing cytotoxic granzyme B (GZMB+) γδ T cells δ TCR (dTCR+) infiltrating the tumor parenchyma, marked by PanCK, as indicated by arrows. Scale bar, 50 microns.

Close modal

γδ T cells have three developmentally distinct cell types distinguished by the choice of Vδ segment during VDJ recombination (31). Using our γδ-specific TCR enrichment protocol, we found that Vδ1 cells in MCCs preferentially paired with Vγ3 and Vγ4 chains, and Vδ2 cells predominantly paired with the Vγ9 chain. We identified nine clusters of γδ cells: seven Vδ1 and two Vδ2, with few Vδ3 cells primarily found in the Vδ1 clusters (Fig. 5B–D).

The predominant γδ cells among the MCC TILs were Vδ1 cells, consistent with previous reports (Fig. 5D; ref. 32). Similar to CD8 Trm cells in MCC, Vδ1 and Vδ3 cells are tissue resident. In contrast, Vδ2 cells are the primary γδ cell in the circulation. Vδ1 and Vδ3 cells (but not Vδ2 cells) appeared to undergo antigen-dependent activation. Vδ1 cells expressed multiple markers of co-inhibition (LAG3, PDCD1, TIM3), activation (41BB, KLRC2), and effector functions (GZMA, GZMB, FASLG), as well as multiple inhibitory killer cell immunoglobulin-like receptors (KIR; KIR2DL1, KIR2DL3), and transcription factors associated with CD8 exhaustion (EOMES, BATF) or tissue residence (RUNX3). Many of these were significantly enriched compared to those in Vδ2 cells (Supplementary Tables S26–S28; Fig. 5E–G). Conversely, Vδ2 cells showed high expression of circulatory markers (CCR7, CD127, CD62L) along with higher expression of AP-1 family members, thus broadly matching the expression patterns observed in CD8 Tcirc cells (Fig. 3D).

Using CITE-seq antibody-based analysis, tumor-resident Vδ1 cells expressed less surface CD3 compared to circulating Vδ1 cells and/or Vδ2 cells in the blood or tumor, reflective of the receptor downregulation following activation (Supplementary Table S28; Fig. 5F). For Vδ1 cells, there were 397 unique clonotypes, of which 128 (32.2%) were expanded, suggesting higher clonal proliferation and clonal focusing than in Vδ2 cells (372 total clones, 85 expanded). The median clonal size was significantly higher among Vδ1 (17 vs. 3; P < 2.2e−16, Wilcoxon test; Supplementary Fig. S16A). Almost all the expanded TCRs from 128 clones had biochemically similar δ-chain CDR3s (see “Methods”), suggesting that they responded in a clonotype-specific manner to a public antigen, whereas the rest of the Vδ1 CDR3s were not expanded and were not similar, suggesting a diverse background of TCRs that may not recognize the same antigen (Fig. 5H).

Pseudotime analyses suggest that Vδ1 cells start from a naïve-like population (cluster C08_GD), which consisted of T-cell clones that did not proliferate (as determined by clonotype analysis) and enriched for the expression of markers associated with naïve γδ T cells, including CD27, CD28, CD45RA, CD127, CCR7, and TCF7 (Supplementary Fig. S16B; ref.33). The Vδ1 trajectory shifted primarily toward clusters C02_GD, C15_GD, and C10_GD, matching the clonal expansion observed previously (Fig. 5I; Supplementary Fig. S16C). These differentiated cells had higher levels of cell cycle markers (MKI67), exhaustion markers (ENTPD1, LAG3, HAVCR2, TIGIT, BATF), cytotoxic effectors (GZMH and GZMB), and cell migration proteins (CXCR6 and VCAM1; Supplementary Table S29; Fig. 5J). These data suggest that Vδ1 cells are CD8 T cell-like, in that they differentiate to a chronically activated phenotype in response to a CDR3-dependent antigen (Fig. 3D). Unfortunately, there were insufficient cells to perform pseudotime analysis of Vδ2 or Vδ3 cells.

To orthogonally validate the functional relevance of γδ T cells in MCCs, we performed mIF on seven patients (eight samples), including four with RNA-seq signatures suggestive of γδ T–cell infiltration. For a subset of MCCs, γδ cells were also detected in the tumor parenchyma expressing cytotoxic markers (Granzyme B; Fig. 5K). Interestingly, we found no statistically significant differences in Vδ1 infiltration between skin and LN samples in either our single-cell data or bulk RNA-seq data. All highly enriched Vδ1 samples were found in the skin, but the numbers of samples preclude statistical significance (Supplementary Fig. S13).

Spatial Transcriptomics Localizes Key Immune Signatures and a Cellular Triad Associated with Response

To resolve the TME spatially, we first used the GeoMx Digital Spatial Profiler, which identified 180 regions of interest (ROI) across 15 samples from 12 patients. These 12 patients included four complete responders to ICB after biopsy and eight nonresponders. The varied ROIs allowed for a comprehensive overview of the tissue landscape (Supplementary Figs. S17A, S17B, and S18A).

Differential expression analysis (DE-seq) was performed on these ROIs. Within the tumor region of complete responders (CR), we observed both T-cell genes (LAG3, GZMB, TRDC) and interferon response genes (GBP4), while in nonresponders (all with progressive disease), there was prominent expression of tumor markers (keratins, CD44, HOX genes) and proliferation markers (TYMS, MKI67; Supplementary Table S30; Supplementary Fig. S18B).

We detected a strong IFNγ response signature (GBP4, HLA-I, TAP, and proteasome genes) in the stroma of CR samples. Additionally, T-cell markers (CD27 and CD7), B-cell markers (CD79A and POU2AF1), and macrophage markers (C1QB, CD68, CD163) were identified, confirming the localization of these cells in the stroma. Conversely, in progressive disease samples, we identified tumor markers (keratins, PRAME) and proliferation markers (TYMS, MCM, E2F1; Supplementary Table S31; Supplementary Fig. S18B). We utilized the Trm and Tcirc gene signatures that we had already developed to spatially examine these specific T-cell lineages. We observed that while both Trm and Tcirc were upregulated in the stroma of responders, only Trm was upregulated in the tumors of responders. Nonresponders failed to show enrichment of Trm or Tcirc genes, regardless of the location (Fig. 6A).

Figure 6.

Spatial and temporal analysis of TILs. A, Violin plots of the relative expression of GeoMx ROIs of Trm/Tcirc signature genes (derived from our scRNA-seq dataset) by module score in the stroma or tumor parenchyma. B, UMAP projection of 614,209 cells segmented using the CosMx assay. The cells are colored according to the inferred cell type. Cells were clustered in Seurat and batch-corrected using SCTransform. C, Unsupervised hierarchical clustering heatmap of cell type colocalization as measured by the DoC within CosMx fields of view (FOV). Neighboring cells were identified within a 54 micron radius of each cell, and then the DoC was determined using the SpatialTIME (34) method. A high DoC was observed among T cells, B cells, DCs, and pDCs, is outlined in the top (left). Ave, average; DC, dendritic cells; Endo, endothelial cells; Epi, epithelial cells; Fibro, fibroblasts; Mac, Macrophages; Mono, monocytes; pDC, plasmacytoid dendritic cells; Tumor cyc, tumor cycling cells. D, A sample FOV showing colocalization of T cells, B cells, and pDC at the tumor–stroma interface using CosMx cell segmentation representation (left) and morphological markers showing para-nuclear dot-like expression of Pan cytokeratins (CK), with high CD298 expression across immune cells. Scale bar, 50 µm. E, Bar charts of the cell types found in T-cell neighborhoods (54 µm radius) showing prevalence of pDC, DC, and B cells in T-cell neighborhoods of responders (R) vs. T-cell neighborhoods of nonresponders (NR). Comparisons were made using the χ2 test, ****, P value < 2.2e−16. F, A dot plot of receptor–ligand interactions from CellChat on the scRNA-seq dataset identifies multiple activating chemokines, cytokines, and surface receptor interactions between B cells, DCs, and pDCs to exhaustion, CD8 Trm cells (CD8 Tex.). G, Differential gene expression analysis (DE-seq2) comparing matched pre- vs. post-ICB sample pairs across seven nonresponders and seven responders shows upregulation of inflammatory genes after ICB treatment. Among responders, 1,560 genes were upregulated and 995 genes were downregulated after therapy. (DE-Seq2, Padj. < 0.1). Only two differentially expressed genes were identified among the nonresponders. H, Enrichr gene list analysis of DE genes in pre- vs. post-ICB matched pairs from responders shows inflammatory pathways upregulated and cell cycle pathways downregulated after treatment. Only protein-coding genes with FDR < 0.1 were included in the analysis. The top five enriched gene sets from the Hallmark gene set libraries are shown for the DE genes. Gene sets were ranked based on the Enrichr combined score, which is a measure of both Fisher’s exact test P value and the z-score of the deviation from the expected rank of the gene set. I, Corrected cellular proportions of Trm cells in 15 post-ICB treated samples show CD8 Trm infiltration is higher (3.01-fold) in partial responders (PR) vs. progressive disease (PD) after treatment. Samples are split by response prior to biopsy. Five of the samples represent a new cohort of post-ICB samples used to verify results from the original scRNA-seq dataset. Wilcoxon test. J, Violin plots of CD8 cells in MO_MCC_079 and MO_MCC_085, two scRNA-seq pre/post sample pairs. Exhaustion score expression was split by treatment timeline and grouped by clonotype expansion showed that expanded cells were more likely to be exhausted pretreatment, with exhaustion levels significantly decreased after treatment. The exhaustion score (37) was derived from Seurat AddModuleScore.

Figure 6.

Spatial and temporal analysis of TILs. A, Violin plots of the relative expression of GeoMx ROIs of Trm/Tcirc signature genes (derived from our scRNA-seq dataset) by module score in the stroma or tumor parenchyma. B, UMAP projection of 614,209 cells segmented using the CosMx assay. The cells are colored according to the inferred cell type. Cells were clustered in Seurat and batch-corrected using SCTransform. C, Unsupervised hierarchical clustering heatmap of cell type colocalization as measured by the DoC within CosMx fields of view (FOV). Neighboring cells were identified within a 54 micron radius of each cell, and then the DoC was determined using the SpatialTIME (34) method. A high DoC was observed among T cells, B cells, DCs, and pDCs, is outlined in the top (left). Ave, average; DC, dendritic cells; Endo, endothelial cells; Epi, epithelial cells; Fibro, fibroblasts; Mac, Macrophages; Mono, monocytes; pDC, plasmacytoid dendritic cells; Tumor cyc, tumor cycling cells. D, A sample FOV showing colocalization of T cells, B cells, and pDC at the tumor–stroma interface using CosMx cell segmentation representation (left) and morphological markers showing para-nuclear dot-like expression of Pan cytokeratins (CK), with high CD298 expression across immune cells. Scale bar, 50 µm. E, Bar charts of the cell types found in T-cell neighborhoods (54 µm radius) showing prevalence of pDC, DC, and B cells in T-cell neighborhoods of responders (R) vs. T-cell neighborhoods of nonresponders (NR). Comparisons were made using the χ2 test, ****, P value < 2.2e−16. F, A dot plot of receptor–ligand interactions from CellChat on the scRNA-seq dataset identifies multiple activating chemokines, cytokines, and surface receptor interactions between B cells, DCs, and pDCs to exhaustion, CD8 Trm cells (CD8 Tex.). G, Differential gene expression analysis (DE-seq2) comparing matched pre- vs. post-ICB sample pairs across seven nonresponders and seven responders shows upregulation of inflammatory genes after ICB treatment. Among responders, 1,560 genes were upregulated and 995 genes were downregulated after therapy. (DE-Seq2, Padj. < 0.1). Only two differentially expressed genes were identified among the nonresponders. H, Enrichr gene list analysis of DE genes in pre- vs. post-ICB matched pairs from responders shows inflammatory pathways upregulated and cell cycle pathways downregulated after treatment. Only protein-coding genes with FDR < 0.1 were included in the analysis. The top five enriched gene sets from the Hallmark gene set libraries are shown for the DE genes. Gene sets were ranked based on the Enrichr combined score, which is a measure of both Fisher’s exact test P value and the z-score of the deviation from the expected rank of the gene set. I, Corrected cellular proportions of Trm cells in 15 post-ICB treated samples show CD8 Trm infiltration is higher (3.01-fold) in partial responders (PR) vs. progressive disease (PD) after treatment. Samples are split by response prior to biopsy. Five of the samples represent a new cohort of post-ICB samples used to verify results from the original scRNA-seq dataset. Wilcoxon test. J, Violin plots of CD8 cells in MO_MCC_079 and MO_MCC_085, two scRNA-seq pre/post sample pairs. Exhaustion score expression was split by treatment timeline and grouped by clonotype expansion showed that expanded cells were more likely to be exhausted pretreatment, with exhaustion levels significantly decreased after treatment. The exhaustion score (37) was derived from Seurat AddModuleScore.

Close modal

Next, for single-cell resolution, we performed CosMx Spatial Molecular Imager analysis. We selected 122 fields of view (FOV) sized 0.9 × 0.7 mm across the tumor–stromal interface from 13 samples across 12 patients (six responders, four nonresponders). We segmented 614,209 cells (Supplementary Table S32; Supplementary Fig. S19A–S19D; Fig. 6B; “Methods”). Next, we deployed SpatialTIME, a permutation-based method that we developed to generate a degree of clustering (DoC) score to rank whether cell populations are more colocalized versus more dispersed than a distribution based on complete spatial randomness across all possible binary comparisons at specified neighborhood radii (34). We then used the presence of elevated DoC to identify key cellular immune circuits associated with ICB response. To do this, we performed unsupervised hierarchical clustering to identify four cell types (T cells, B cells, pDC, and DC) exhibiting high DoC across all FOVs and the entire range of radii r = 250 to 500 (90–180 µm diameter; Supplementary Table S33; Supplementary Fig. S20A–S20D; Fig. 6C). The highest DoC corresponding to the highest levels of colocalization (>4.5 × 105) was seen across two circuits: T cell–B cell–DC and T cell–B cell–pDC. An example of this is seen in an FOV highlighting T cells, B cells, and pDC all colocalized at the tumor–stroma interface (Fig. 6D). We further sought to identify whether these distributions differed between responders and nonresponders. Among responders, B cells, DCs, and pDCs showed 61.9-, 1.8-, and 14.3-fold higher counts, respectively, with a r = 300 (108 µm diameter) in the neighborhood around T cells (P < 2.2e−16, χ2; Fig. 6E; Supplementary Fig. S20E). Conversely, T cells were preferentially colocalized with 1.7- and 13.5-fold more tumor and tumor cycling cells, respectively, in the same sized neighborhoods (r = 300; 108 µm diameter), suggesting a paucity of intact immune circuits in nonresponders (Fig. 6E; Supplementary Fig. S20E). We interpret this to suggest that the presence of a key immunological triad of B cells, T cells, and DCs or pDCs is correlated with ICB response.

We then leveraged the droplet-based scRNA-seq dataset to identify receptor-ligand interactions because of higher numbers of transcripts per gene, focusing on B cell and myeloid populations (including dendritic cell subsets; Supplementary Fig. S6A). Using CellChat (35), we found that B cells, DCs, and pDCs interacted with Trm T cells via multiple chemokine–chemokine receptor interactions (i.e., CXCL16 to CXCR6), costimulatory ligand–receptor interactions (i.e., 41BBL (TNFSF9) to 41BB (TNFRSF9), CD80/CD86 to CD28), and activating cytokines [i.e., IL15 to IL15 receptor, IFNA2/IFNW1 to IFNAR1/2 (IFNα receptor); Supplementary Table S34; Fig. 6F].

ICB Increases Recruitment of Trm Cells and Multiple Inflammatory Pathways in Responders

To examine the effects of ICB, we examined a cohort of seven responders and seven nonresponders from which we sampled matched tumor tissue pre- and post-therapy. Among responders, 1,560 genes were upregulated and 995 downregulated after therapy. (DE-Seq2, Padj. < 0.1; Supplementary Table S35). In contrast, only two differentially expressed genes were identified among nonresponders (Fig. 6G).

Overall, responders showed upregulation (1.34–1.44 fold; P < 0.05) of multiple inflammatory signatures after treatment including our Vδ1 signature and an IFNG signature (10). There was a strong overlap in the pathways and the genes upregulated in pre-ICB responders and those upregulated in responders post-ICB (Fig. 6H; Supplementary Table S36; Supplementary Fig. S21A and S21B). These include markers of B cells (e.g., immunoglobulins), pDC/DC markers (e.g., PTGDS), T-cell markers (e.g., CD3, TCR), chemokines/cytokines (e.g., IFNG, IL15, CXCL16), CD8 Trm markers (e.g., CD69, PRF1, CD8B, CXCR6), and multiple interferon response genes (e.g., HLA, IRF1, IFI16) and gene signatures of Trm cells (but not Tcirc cells; 1.44-fold; P = 0.031; Wilcoxon; Supplementary Fig. S21B). These data collectively suggest that ICB increases the recruitment or expansion of immune cells that were present prior to ICB. Cell cycle and cell proliferation pathways were downregulated in responders post treatment. These included markers of tumor proliferation (FOXM1, TOP2A, MKI67, E2F7) and MCC-specific markers (e.g., ATOH1, NEFM), suggesting tumor clearance. In contrast, nonresponders show little to no change after ICB treatment (2%–9% change, P > 0.5) and were thus lower than temporally matched responders pre- or post-ICB (Supplementary Fig. S21).

Next, we examined a cohort of scRNA-seq samples only acquired from patients after immunotherapy. These 15 samples included five additional samples that were projected on the original single-cell dataset (see “Methods”). Consistent with our bulk RNA-seq data, responders (n = 5) had 11.1-fold lower levels of tumor cycling cells and 3.01-fold higher CD8 Trm cells (3.01-fold increase) following ICB (P < 0.05, Wilcoxon; Fig. 6I; Supplementary Fig. S21C).

Next, we examined antigen receptor sequences to study clonal dynamics in the TME after ICB in our bulk RNA-seq data using MIXCR (36). As suggested in our differential expression analysis, we observed 60- to 400-fold more total TCR or BCR clones in responders versus nonresponders (Wilcoxon; P < 0.05). Nonresponders had no expanded TCRs and BCRs prior to treatment, while 3/7 responders had expanded TCR clones and 5/7 responders had expanded BCR clones prior to treatment. After treatment, 1/7 (TCR) and 2/7 (BCR) nonresponder samples showed evidence of new, expanded clones. In contrast, all of the responders had expansion of newly recruited and preexisting TCR and BCR clones (Supplementary Fig. S22A–S22D, see “Methods”). Thus, both preexisting immunity and the recruitment of new, reactive T cells and B cells are implicated in response.

As MIXCR data can be biased by differences in TCR mRNA between cells, we used our matched single-cell data to more precisely assess CD8 T-cell dynamics. We examined two paired sets of ICB responder samples with pre- and post-therapy scRNA-seq from both the blood and tissue. One sample pair had matched bulk RNA-seq and one pair was new. After treatment, we saw the same results as our MIXCR data, where both samples had expansion of preexisting clones, but also recruitment of expanded clones (Supplementary Fig. S23A and S23B). Expanded cells from both patients were enriched for putative Trm-like signatures. They expressed CD103 and exhaustion markers [exhaustion score (37), PD-1, and LAG3]. Following anti-PD1 therapy, expanded cells had significantly lower levels of exhaustion markers, including a reduced exhaustion score and a 2.11 to 5.44-fold decrease in PD-1 and LAG3 expression, respectively (P < 1e6; Wilcoxon; Fig. 6J; Supplementary Fig. S23C), which may reflect increased functionality after ICB. Full cell type analysis of pre-post matched scRNA-seq from the blood (21 pairs) did not reveal any significant cell clusters correlated with response or resistance. Tumor-specific T cells comprised a maximum of 0.5% of the circulating T cells, but the number of cells per sample was not high enough to reliably capture a cluster of those cells (38).

In this study, we compiled the largest database to date of MCC tumor samples and matched blood with multimodal profiling of a total of 116 patients. The breadth and depth of our studies have allowed us to confirm findings across multiple orthogonal datasets, highlight the importance of tumor sampling (over blood), and identify multiple novel candidate biomarkers that may be clinically actionable. For instance, the negative impact of tumor cell proliferation on ICB response (Figs. 1F, G, 2H, and I) may be validated in CLIA settings using assays assessing Ki-67 expression. Moreover, next-generation sequencing may similarly enable prospective validation of IFNγ and/or IFNα signatures to predict responses to immunotherapy (Figs. 1D, E, 2D, and F; ref. 10).

Our data emphasize the role of tissue-resident lymphocytes in MCCs. Despite representing a minority (∼45%) of the total CD8 T cells, tissue-resident CD8 T cells appear to be the primary tumor-specific population responsible for tumor recognition and killing. Moreover, the prevalence of this tissue-resident population (but not the circulating T-cell population) is clinically important. In contrast to some other cancer types (39), their presence in the tumor prior to immunotherapy predicted responses (Fig. 4H and I).

Our study provides novel insights into γδ T cells in the TME. While others have reported significant infiltrates of γδ T cells (32), our modified ECCITE-seq allowed unique γδ TCR sequencing with protein markers, which enables linkage of VDJ, transcriptome, and proteomics. This analysis indicated that skin-resident Vγ3Vδ1 and Vγ4Vδ1 cells were the tumor-reactive population (and not the circulating Vγ9Vδ2 cells; Fig. 5E–J).

Vδ1 cells in MCCs expressed costimulatory and inhibitory receptors commonly expressed by innate NK cells including killer cell lectin-like receptor (KLR)/KIR family members (Fig. 5E and J). However, our analysis highlighted deep commonalities with CD8 Trm cells. Like CD8 Trm cells, they express acute activation markers (such as 41BB), undergo clonal expansion, and acquire markers associated with T-cell exhaustion (e.g., PD-1, LAG3, TIGIT). The pseudotime analyses paralleled those of CD8 Trm cells, wherein they both started in a TCF7+ cell state and ended in an exhaustion state and loss of BTG1, a transcriptional repressor of lymphocyte effector function (Fig. 5J; ref. 22). Taken together, these skin-resident Vδ1 cells appear to display a form of functional convergence with skin-resident CD8 Trm cells in driving antitumor immunity (Supplementary Fig. S24A and S24B).

Consistent with the recent reports, these Vδ1 cells are enriched in tumors with altered MHC-I expression (40). In contrast to previous reports, our TCR sequencing data suggest that the CDR3 region is critically important for Vδ1 cells (Figs. 4D and 5I; refs. 40, 41). Through single-cell γδ CDR3 sequencing, we demonstrated clonal expansion of Vδ1 cells that express biochemically similar CDR3s, highlighting the possibility of shared reactivity to a common (as yet unidentified) antigen (Fig. 5I).

Importantly, multiple spatial approaches suggest that T cells in the MCC TME do not function independently. Our data highlights the importance of cellular neighborhoods in response to immunotherapy (Fig. 6A–E).

Finally, we identified novel biomarkers, such as IL1, type I interferon and neural stem cell markers. Both IL1 and type I interferon have been described as having opposing protumor and antitumor roles in cancer (4244). However, our data suggest that exogenous type I interferon and/or IL1 blockade may enhance immunotherapy efficacy. OTX2 and other neural stem cell markers may represent novel targets to improve response to ICB in MCC (Supplementary Fig. S25). MCCs may have unique, yet undiscovered, properties that highlight the biological importance of these molecules, specifically for this cancer type. Alternatively, this unique genomically homogeneous dataset (particularly VP-MCCs) may provide statistical power to make discoveries that are not possible for genetically heterogeneous cancers of other types. In other cancer types, these signals may be subsumed, for example, by the tumor mutation burden, which is required to make the tumor immunogenic. Thus, we believe that many of these results are applicable to other cancers, especially skin cancers.

Patient Selection and Response Assessment

A total of 116 patients with MCC treated at three institutions (Moffitt Cancer Center, Northwestern University Lurie Cancer Center, University of Washington) were included for analysis. The cohort was 79% male, median age 73 years (range, 18–93 years). Disease stage and immunocompromised status was reported in Table 1 and Supplementary Table S1. Inclusion criteria were histologically confirmed diagnoses of MCC made within the Department of Pathology at the corresponding institution and treatment with systemic immunotherapy. The clinical response to ICB was defined as complete response, partial response, stable disease, or progressive disease. However, for the purposes of all molecular analyses, characterization of clinical response to ICB was dichotomized to responder (complete and partial response) versus nonresponder (stable and progressive disease) status. These assessments were made on manual review of all electronic medical records on the basis of a combination of histopathologic evaluation (for neoadjuvant therapy), clinical assessment (clinical assessment of skin and/or lymphadenopathy), and serial radiographic assessment (computed tomography or positron emission tomography—computed tomography).

Complete responses were defined as complete radiographic and clinical response, or, where applicable, concordant pathological complete response. In the case of adjuvant immunotherapy, complete responses were noted in situations where surgery with curative intent had evaluable clinical or radiographic residual disease burden that was subsequently treated with ICB with clearance of disease. Any intermediate responses with residual disease burden were considered partial responses.

Progressive disease was defined as enlargement of overall burden of disease as noted by a combination of clinical and radiological assessment and/or the emergence of new metastases. In the case of adjuvant ICB administered following surgery with or without radiation, any recurrence or progression was considered progressive disease (n = 3). One patient on adjuvant ICB had stable disease with neither disease resolution nor progression. One mixed response was noted and considered an overall partial response in terms of overall tumor burden but the lesion used for molecular analysis showed no evidence of response to therapy and was considered as a progressive disease (MO_MCC_057). Death or date of last follow up was the clinical endpoint used for survival analysis.

Sample Collection

Fresh tissue and blood specimens were collected from patients with MCC treated and managed at the H. Lee Moffitt Cancer Center Department of Cutaneous Oncology following written informed consent under the IRB-approved Total Cancer Care Protocol (MCC 14690, 94962) and performed in accordance with the Declaration of Helsinki. Collected specimens were processed and used under IRB-approved MCC Protocol 19191. Freshly collected tumor specimens were dissected for single-cell dissociation, formalin fixation with subsequent paraffin embedding, or flash frozen. Additional archival tissues at Moffitt were processed under waiver of consent (MCC 19191).

At Northwestern, fresh samples were provided by the Skin Biology & Diseases Resource-Based Center Translational & Experimental Skin Testing & Immune Tracing Core and were obtained through the IRB-approved Dermatology Tissue Acquisition and Repository (IRB # STU9443) or through the IRB-approved Dermatopathology Tissue Repository for Research (IRB # STU24696) and performed in accordance with the Declaration of Helsinki. Freshly collected tumor specimens were dissected for single-cell dissociation, formalin fixation with subsequent paraffin embedding, or flash frozen.

Blood was collected via phlebotomy following informed consent as above and PBMCs isolated under Ficoll gradient and cryopreserved in ∼1 to 2e6 cell aliquots.

scRNA-seq Tissue Sample Processing

Tissue Dissociation

Human skin tumor biopsies or portions of surgical resections were dissociated into single-cell suspensions using either the Miltenyi Biotec human tumor dissociation kit (enzymes H, R, A) or a mixture of RPMI, DNAse (0.1 mg/mL), collagenase IV (2 mg/mL), and hyaluronidase (0.1 mg/mL). Biopsies were gently minced into 1 to 2 mm pieces, transferred to an enzyme mix as above, and incubated at 37°C for 30 to 60 minutes at 800 rpm in a thermomixer or by gentle rocking. Following incubation, cells were filtered, washed, and cryopreserved.

Cryopreserved, digested MCC tumor samples were thawed and processed following a modified version of the 10× Demonstrated Protocol “Thawing Dissociated Tumor Cells for Single Cell RNA Sequencing” and BioLegend TotalSeq C protocol. The samples were thawed, labeled with TotalSeq C antibodies, and stained with 7-AAD viability staining solution and Apotracker Green (BioLegend). Cells were then subjected to fluorescence-activated cell sorting (FACS) to sort the CD45+ and CD45 cell populations for target enrichment of 2/3 CD45+ cells. After sorting, the final cell preparation was resuspended in RPMI + 10% FBS + SUPERase•In RNase Inhibitor (Sigma), filtered, and counted to ensure an optimal concentration of 1e6 cells/mL. Finally, the cell suspension was processed for encapsulation and reverse transcription using the 10× 5′ v2 Chromium platform, according to the manufacturer’s instructions.

scRNA-seq PBMC Sample Processing

Cryopreserved PBMCs were thawed, labeled, and prepared for 10× droplet encapsulation following a modified protocol adapted from the 10× and BioLegend TotalSeq C protocols. Cells were blocked using Human TruStain FcX Fc blocking reagent (BioLegend) and stained with TotalSeq C and hashing antibodies (BioLegend). Live cells were sorted using FACS with a gate set for CD45+ and CD3+ cells. The sorted cells were combined with CD3 cells to maintain a 1:3 ratio and cryopreserved in Bambacker preservation medium with SUPERase•In RNase Inhibitor (Sigma). Equal volumes of four to five hashed samples were combined to ensure that samples stained with the same hashing antibody were not mixed. The cells were slow-frozen at −80°C before being transferred to liquid nitrogen for long-term storage. Thawing and final sample preparation for encapsulation were performed immediately before loading onto the 10× 5′ v2 Chromium platform. Sequencing parameters were set for the 5′ cDNA, αβ VDJ, γδ VDJ, and ADT libraries according to the targeted cell number for scRNA-seq.

TCR Sequencing

αβ/γδ TCR libraries were prepared following the 10× Genomics Single Cell V(D)J protocol. TCR γδ libraries were prepared following the 10× Genomics protocol to generate TCR αβ libraries with modifications. Specific primers were designed, including R1_hTRDC (5′AGCTTGACAGCATTGTACTTCC), R1_hTRGC (5′TGTGTCGTTAGTCTTCATGGTGTTCC), R2_hTRDC (5′TCCTTCACCAGACAAGCGAC), R2_hTRGC (5′GATCCCAGAATCGTGTTGCTC), and the forward primer (5′GATCTACACTCTTTCCCTACACGACGC), which were resuspended in 100 µmol/L EB buffer. The library preparation involved two PCR reactions: PCR1, using Human γ/δ mix 1 (R1_hTRDC + R1_hTRGC), and PCR2, using Human γ/δ mix 2 (R2_hTRDC + R2_hTRGC), substituting the primers as detailed. PCR1 was performed using 2 to 5 µL full-length cDNA, 50 µL 2× Kapa HIFI Master Mix, 5 µL cDNA additive, 2 µL 20 µmol/L forward primer, and 2 µL 20 µmol/L human γ/δ mix 1, with a reaction volume of 100 µL using water. The PCR was performed for 14 cycles. PCR2 was performed with 35 µL bead elution, 50 µL 2× Kapa HIFI Master Mix, 5 µL cDNA additive, 2 µL 20 µmol/L forward primer, and 2 µL 20 µmol/L human γ/δ mix 2, with a reaction volume of 100 µL using water. The PCR was performed for 12 cycles. Cleanups and PCR conditions were identical to the 10× protocol but γ/δ libraries required additional amplification cycles because of the rarity of γδ T cells compared to αβ T cells in most cell populations. Finally, TCR γ/δ-enriched libraries were processed according to the 10× Genomics Single Cell V(D)J protocol.

FFPE RNA-seq

Formalin-fixed, paraffin-embedded (FFPE) tissue blocks were sectioned at a thickness of 5 μm. RNA was extracted from FFPE tissue sections using the RNeasy FFPE Kit (Qiagen) according to the manufacturer’s instructions. The extracted RNA was treated with an RNase-Free DNase Set (Qiagen) to remove any genomic DNA contamination. RNA quality and quantity were assessed using a Bioanalyzer (Agilent) and Qubit fluorometer (Thermo Fisher Scientific), respectively.

RNA sequencing libraries were prepared using the TruSeq Stranded mRNA LT Sample Prep Kit (Illumina), according to the manufacturer’s instructions. Briefly, 500 ng total RNA was used for poly(A) mRNA selection, fragmentation, and cDNA synthesis. The resulting cDNA was end-repaired, adenylated, and ligated using Illumina-specific adapters. PCR amplification was performed to enrich adapter-ligated fragments. The final library was purified and validated for size distribution using a Bioanalyzer (Agilent) and quantified by qPCR using a Kapa Library Quantification Kit (Kapa Biosystems). Libraries were pooled at equimolar concentrations and sequenced using an Illumina NovaSeq 6000.

Fluorescent Multiplex Immunofluorescence Panel Procedure

FFPE tissue samples were immunostained using the AKOYA Biosciences OPAL TM 7-Color Automation IHC kit (Waltham) on a BOND RX autostainer (Leica Biosystems). The OPAL 7 color kit uses tyramide signal amplification (TSA) conjugated to individual fluorophores to detect various targets within the multiplex assay.

The sections were baked at 65°C for 1 hour then transferred to BOND RX (Leica Biosystems). All subsequent steps (deparaffinization and antigen retrieval) were performed using an automated OPAL IHC procedure (AKOYA). OPAL staining of each antigen was performed as follows: heat-induced epitope retrieval (HIER) was achieved with EDTA buffer (pH 9.0) for 20 minutes at 95°C before the slides were blocked with AKOYA blocking buffer for 10 minutes. The slides were then incubated with the primary antibody, GZMB (CST, D6E9W, RRID:AB_2799313, 1:75, dye 570) at RT for 30 minutes, followed by incubation with OPAL HRP polymer and one of the OPAL fluorophores during the final TSA step. Individual antibody complexes were stripped after each round of antigen detection in an optimized order and were determined independently for each panel. After the final stripping step, DAPI counterstain was applied to the multiplexed slide and removed from the BOND RX for coverslipping with ProLong Diamond Antifade Mountant (Thermo Fisher Scientific). All slides were imaged using the Vectra3 Automated Quantitative Pathology Imaging System.

Quantitative Image Analysis

Multilayer TIFF images were exported from InForm (AKOYA) and loaded into HALO (Indica Labs) for quantitative image analysis. A classifier was trained to identify areas of the tumor, stroma, or non-tissue regions. Pan-cytokeratin was used to train tumor regions because it is a masking marker for tumor cells. A classifier was created and tested on various images of the image set. The tissue was segmented into individual cells using the DAPI marker, which stains the cell nuclei. For each marker, a positivity threshold within the nucleus or cytoplasm was determined per marker based on published staining patterns and intensities for that specific antibody. After setting a positive fluorescent threshold for each staining marker, the entire image set was analyzed using the created algorithm. The generated data included positive cell counts for each fluorescent marker in the cytoplasm or nucleus and the percentage of cells positive for the marker. The data was sorted by classification to include the entire tissue, stroma, or tumor regions. Along with the summary output, per-cell analysis was exported to provide the marker status, classification, and fluorescence intensities of every individual cell within an image.

Local density analysis was performed by outlining the bulk tumor based on para-nuclear dot-like accentuation of panCK expression. Epidermal/follicular expression of panCK was excluded from analysis. Following the definition of the tumor-stromal interface, bands of 40 µm widths were defined as +500 microns (out of tumor into stroma) and −500 microns (into tumor interior), and the local density of cell counts according to marker expression was calculated per mm2.

Spatial Transcriptomics Nanostring GeoMx

Tissue slices of 15 MCC tumor samples from 12 patients were placed on charged glass slides and stained with four immunofluorescent markers to assist in the selection of 12 ROIs per sample. The tissue was selected from a cohort of patients with MCC who received immunotherapy (four responders and eight nonresponders). FFPE human skin samples were baked for 1 hour at 65°C and processed on the Leica automation platform in three major steps:(i) slide baking and deparaffinization, (ii) antigen retrieval for 20 minutes at 100°C, (iii) 1.0 μg/mL Proteinase K treatment for 15 minutes. Slides were incubated overnight with GeoMx CTA or WTA assay probe cocktail. The following day, the slides were washed and subjected to morphological marker incubation before loading onto a GeoMx machine for fluorescence scanning and ROI selection. Barcodes were collected and the subsequent barcoding reading was completed on the Illumina NGS platform.

The markers used in the ROI selection were SYTO13 (DNA/nuclei), KRT18 (keratin/cancer cells), PTPRC (CD45/lymphocytes), and CD3E (T cells). Reagent: Syto13; Manufacturer: Thermo Fisher; ID: 121301310. Protein: PanCK; Manufacturer: Novus; Clone: AE1/AE3; ID: NBP2-29429, RRID:AB_3068002. Protein: CD45; Manufacturer: CST; Clone: D9M8I; ID: 13917BF, RRID:AB_2750898. Protein: CD3; Manufacturer: Origene; Clone: UMAB54; ID: UM500048, RRID:AB_2629062.

The classifications assigned to the ROIs were based on manual examination by the pathologist in our team, aided by DAPI and PanCK immunofluorescent stain, as presented in Supplementary Fig. S18A. Selection of the ROIs was done by the pathologist before RNA quantitation by GeoMx. Those ROIs in which the pathologist observed a predominance of epithelial cells were classified as tumor ROIs. Contrarily, those ROIs showing the absence of epithelial cells were classified as stroma. Variations of these ROIs were also included, in which tumor or stroma ROIs could also present infiltration of immune cells (CD45 or CD3 stain). ROIs of arbitrary shapes were selected to represent the tumor and stroma, including areas close to the tumor–stromal interface and central tumor areas corresponding to at least 150 cells, assigned to five categories with respect to their spatial location and level of immune infiltration:(i) “immune cold” stroma (paucity of immune cells), (ii) immune-excluded stroma (immune cells distant from tumor nodule), (iii) immune-infiltrated stroma (stroma adjacent to tumor infiltrated by immune cells), (iv) tumor close to immune cells (adjacent tumor areas to immune-infiltrated stroma), and (v) inner tumor (central areas of tumor nodules typically devoid of significant immune infiltrates; Supplementary Fig. S17A). The slides were profiled for gene expression using the Nanostring Cancer Transcriptome Atlas panel (CTA, nine samples/108 ROIs) and Whole Transcriptome Atlas panel (WTA, six samples/72 ROIs). The number of reads per ROI varied from 1.1 × 106 to 142.9 × 106 (Supplementary Fig. S17B).

The resulting .dcc files were processed using the GeoMx Analysis Suite v2.5. Probe counts were filtered using the default values and aggregated at the gene level. Given the technical effects associated with the panel used (WTA or CTA), we applied ComBat correction as implemented in the R package sva. We applied third quartile normalization of the counts, followed by log transformation. Normalized counts were used to detect differentially expressed genes between immune infiltration categories and patient therapy responses within each spatial niche (stroma/tumor). Differential gene expression was performed via pairwise t tests in the scran package, and P values were adjusted for multiple testing using false discovery rate (FDR).

Spatial Transcriptomics CosMx Spatial Molecular Imaging

To study the spatial patterns in gene expression of immune and tumor cells, we profiled six slides containing MCC tissue using the 960-gene Immune Oncology panel Nanostring’s Spatial Molecular Imaging (SMI) platform. Tissue samples were obtained from 12 patients, six of which showed a positive response to therapy, four were nonresponders and two were not accessible. FFPE tissue sections were prepared for CosMx SMI profiling as described by He and colleagues (45). Morphological markers used are as follows.

Protein: CD298; Manufacturer: Abcam; ID: ab167390, RRID:AB_2890241. Protein: B2M; Manufacturer: Abcam; ID: ab214769, RRID:AB_1266996; Protein: PanCK; Manufacturer: Novus; ID: NB2-33200AF532, RRID:AB_2924722. Protein: CD45; Manufacturer: Novus; ID: NBP2-34528AF594, RRID:AB_960384; Protein: CD3; Manufacturer: Origene; ID: UM500048, RRID:AB_2629062.

A total of 122 FOVs were profiled (2–16 per patient), which were chosen by inspecting adjacent hematoxylin and eosin–stained tissue slices (Supplementary Fig. S19A) and reconfirmed by imaging with morphological markers. The raw dataset consisted of 712,617 image-segmented cells, and transcripts falling within the cells were summarized at the gene level. Cells without any recorded transcripts were removed from the downstream analyses (815 cells). The SMI platform includes 19 negative probes for assessment to assist in the removal of background levels for transcript detection and quality control. We removed from the data set cells with more than 5% negative probe transcripts as well as cells with less than 50 expressed genes. To reduce the effect of segmentation artifacts (i.e., two or more cells merged into a single segment—“doublets”), we removed cells with more than 500 expressed genes or 1,250 transcript counts. The filtering procedure resulted in 614,209 cells being used in downstream analyses. We applied SCTransform normalization to the resulting counts. Corrected counts were input to InSituTypeML, a Bayes classifier to transfer the cell types identified in the tissue single-cell dataset to the SMI cells. To account for the background signal, InSituTypeML used the average negative probe count per cell. We also inferred Louvain clusters and tested for differentially expressed genes among clusters using Seurat to manually curate annotations generated by InSituTypeML. We confirmed that DE genes from the assigned CosMx clusters significantly overlapped with the DE genes from the higher resolution single-cell dataset (Supplementary Fig. S19D).

To quantify the degree of dispersion or colocalization of specific cell types within the spatial immune landscape in the MCC samples, we used spatialTIME (34) to measure the DoC across all possible binary combinations of cell subsets based on scRNA-seq data. The method implemented in spatialTIME computes the bivariate Ripley’s K statistic and assumes that cells occur at the same frequency across an FOV (i.e., complete spatial randomness) and is further normalized for area and cell numbers. Because FOVs often show non-uniform cellularity, spatialTIME compares the observed K to the exact K estimate based on a spatial random point process. This difference is the derived measure called the DoC.

MCPyV Epitope Screen

Autologous LCLs were generated using a modified version of the Current Immunology protocol 7.22. PBMCs were thawed and washed with prewarmed complete RPMI medium. After counting and assessing viability, cells were centrifuged and resuspended in complete RPMI with 1.1 µg/mL cyclosporin A concentration of 2.2 × 106 cells/mL. The cell suspension was incubated for 1 hour at 37°C before adding the thawed Epstein-Barr virus (EBV) viral supernatant (ATCC) to achieve a final cell concentration of 2 × 106 cells/mL, with the virus constituting 10% of the final volume. The culture was monitored daily, and when the medium turned yellow, it was gently aspirated and replaced with fresh RPMI medium containing 1 µg/mL cyclosporin A. After 2 to 3 weeks, large cell clumps appeared and cyclosporin was no longer added to the culture. LCLs were maintained at high densities (>1 × 106cells/mL) and thrived in slightly acidic (yellowish) media, with pipetting used to break up large cell clumps, as needed. After expansion, the LCLs were cryopreserved for further use.

The patient’s LCLs were thawed and treated with mitomycin C to inhibit proliferation, followed by washing and plating. Jurkat-76 cells previously transduced with a CD8-NFAT-GFP reporter construct (RRID:Addgene_153417; ref.46) were lentivirally transduced with transgenic TCRs (TWIST Biosciences) and selected in 1 μg/mL puromycin. A total of 116, 13-mer peptides were designed to tile along the small T and large T antigens of MCPyV with 8-mer overlap (Genscript; Supplementary Table S23; ref. 24). Peptides were pooled into 22 pools, such that each peptide was present in two pools. On day 2, the peptide pools were added to the LCLs and incubated for an hour at 37°C. Peptide-loaded cells were washed with cRPMI. Jurkat reporters were added to each LCL well, mixed, and centrifuged before incubation for 20 to 24 hours at 37°C. The next day, the remaining cells were stained with CD69 PE (BioLegend Cat# 310905 (also 310906), RRID:AB_314840), HLA-DR APC-Cy7 (BioLegend Cat# 307618 (also 307617), RRID:AB_493586), CD3 BV421 (BioLegend Cat# 344834 (also 344833), RRID:AB_2565675) antibodies for 30 minutes. The cells were washed, resuspended with 7AAD, and analyzed using a Fortessa HTS flow cytometer to measure Live, HLA-DR-, CD3+ CD69+ GFP + cells. Jurkat-76 cells were received as a gift from Mirjam H. M. Heemskerk on September 25, 2018 (47). They were authenticated as TCR-negative but no additional authentication was performed. Upon initial culture, the cells were periodically tested for mycoplasma with the MycoAlert kit (Lonza). Cultures were passaged up to five times before experiments.

Computational Methods

scRNA-seq Preprocessing

The GRCh38.p13 primary assembly from Ensembl Release 98 and the GENCODE v32 primary assembly were modified as follows. CellRanger 6.1.0 was used to create a reference for scRNA-seq alignment. The MCPyV sequence was then added to the primary and final GTF files.

FASTQ files containing gene expression and feature barcodes (ADT) were aligned to the human hg38 and MCPyV genomes using CellRanger 6.1.0. Ambient RNA was removed using the CellBender pipeline (48). Processed read counts for each sample were imported as Seruat objects, after which TCR-seq data were added. We then filtered the data based on mitochondrial reads (25% cut-off) and minimum RNA genes (200 genes). Doublets were removed using DoubletFinder with the default pN of 0.25 and the optimal pK value calculated with the “find.pK” function. Cells were annotated with ProjectTILS and SingleR using Monaco and HPCA reference datasets. Data were merged into a single Seurat object. We performed standard Seurat (4.0) clustering, Harmony batch correction, and manual cell cluster annotation based on marker genes, including transcription factors, effector molecules, and previously annotated cell types. The data were subclustered by major cell types. Contaminating cells were filtered based on marker gene expression. Differential gene expression, gene enrichment, and GSVA were performed using the default Seurat pipeline, followed by comparison of cell type proportions, analysis of TCR clonotype expression, and TCR linkage analysis.

Enrichr Gene Ontology Analysis

Protein-coding genes were subset upregulated or downregulated genes as indicated and input into the Enrichr pipeline using the enrichR package (3.0) and querying all available databases (9). The resulting enriched gene sets were grouped by database and then sorted by “combined score” which was labeled the Enrichr score.

Pseudobulk Differential Gene Expression Analysis

The cells of interest were subset from the full scRNA-seq object. Cells were then grouped by sample into a pseudobulk matrix using Seurat’s “AggregateExpression” function. Next, this matrix was used to input into differential expression analysis using edgeR (3.36.0) using the “glmLRT” function.

Gene Set Enrichment Analysis

Genes from differential expression analysis were ranked by signed P value. These genes were used as input for the fgsea function from the fgsea package (1.24). Reference genesets were derived from all human geneset in the msigdbr package (7.5.1). To control for cell cycle and tumor cycling cells, GSEA was run on two different pseudobulk DE results on tumor cells. The first result was without any tumor cycling cells and the second was controlling for cell cycling scoring as described in the pseudotime analysis and covariate DE-seq.

Statistical Considerations for RNA-seq Analysis

In the absence of specific hypothesis testing, sample size rationales were not calculated a priori. The major components of data were bulk RNA-seq and scRNA-seq data. These samples numbered 85 (63 for pre-ICB analysis) and 109, respectively. For bulk RNA-seq, primary conclusions were based upon signature based analyses and the power calculation for the RNA-seq experiment was conducted using package “ssizeRNA”. With an assumption of 10,000 tested genes (including the top 100 prognostic genes), a minimum fold change criterion of 2, and accounting for dispersion parameters ranging from 0.1 to 0.4, 63 samples (comprising 32 responders and 31 nonresponders) achieves an estimated probability of more than 80% (desired power) to reject the null hypothesis assuming equality in expression means between the two groups, with an associated FDR of 0.05. Additionally, we performed power calculations based on the gene set–based signature, specifically GSVA scores. Assuming a normal distribution of GSVA scores, our sample size of 63 provides a power of 90% to detect effect sizes (Cohen’s d) at 0.86 and a power >80% to detect effect sizes at 0.75, while assuming a type I error rate of 0.05. In our scRNA-seq experiments, where each sample comprises an average of 5,129 cells. Assuming that each subpopulation contains a minimum of 10 cells, our approach achieves a 95% probability of success to detect cell subpopulations with frequency as rare as 0.003.

Gene Set Module Score

Cells in the scRNA-seq dataset were assigned a module score using the “AddModuleScore” function in the Seurat package. This function was run on the combined Seurat object with all samples. In some cases, the cells were grouped by sample and/or cell type, and the average module score was reported. The following gene sets were used: Hallmark interferon gamma signature (MSIGDB), Hallmark interferon alpha signature (MSIGDB), exhaustion score from the cited paper (37), and NeoTCR8 score from the cited paper (23).

SCENIC Analysis

Following loom file conversion from the subset Seurat object, we executed the standard pySCENIC pipeline (v 0.12.1; ref. 15) in three major steps: gene regulatory network inference, context-dependent enrichments using cis-regulatory motifs, and activity scoring of regulons in individual cells using the AUCell algorithm. Subsequent to the SCENIC analysis, we integrated the results into our Seurat object. We used the FindAllMarkers function, ensuring even sample representation with equal number of cells, to find differentially expressed regulons between responders and nonresponders.

Diffusion Mapping

Cells were reclustered with cell cycle genes regressed using Seurat’s “CellCycleScoring” and “ScaleData” functions. Batch-corrected principal components were input into the DiffusionMap function of the Destiny package (3.14).

Pseudotime Analysis

Cells were subset and reclustered with cell cycle genes regressed using Seurat’s “CellCycleScoring” and “ScaleData” functions. After cell cycle regression, pseudotime was determined using the default pipeline in monocle3 (1.0.0). The “root” node for pseudotime was determine by expression of naïve like markers including CD45RA, TCF7, and non-expression of exhausted or memory markers including PD-1, CD45RO, TOX. Genes were then linearly correlated with pseudotime using Pearson correlation. The next genes were nonlinearly correlated to pseudotime using the “fitGAM” function in the tradeSeq (1.8.0) package. The expression levels of significant genes were scaled across pseudotime and smoothed using the “smooth.spline” function with five degrees of freedom from the stats package (4.1.1). Genes were ordered by maximum value across pseudotime and then smoothed/scaled expression was plotted as a heatmap using the ComplexHeatmap package (2.13.2, RRID:SCR_017270).

CellChat

All cells from the tissue scRNA-seq dataset were made into a pseudobulk average expression Seurat object, grouped by sample and cell type. This was input into the CellChat (1.4.0) pipeline using the human database accessed on April 29, 2023.

Projection of Additional Post-ICB scRNA-seq Samples

Five new post-ICB samples were harmonized using the Harmony package. All cells were mapped to the original Uniform Manifold Approximation and Projection (UMAP) and clusters from the original 49 sample tissue MCC scRNA-seq data using the Seurat “FindTransferAnchors” and “MapQuery” functions.

Single-Cell TCR-seq Preprocessing

FASTQ files for both αβ/γδ VDJ sequencing were input into the Cellranger (3.0.2) vdj pipeline. Both contig annotations and clonotype annotations were added as metadata to the scRNA-seq Seurat objects for downstream analysis.

TCR Linkage Analysis

Identical clonotypes were matched between cell clusters and the likelihood ratio of clonotype matching was calculated for each cluster pair. P-values were determined using Fisher’s test and adjusted using the Benjamini–Hochberg procedure. Finally, log-likelihood ratios were subjected to unsupervised clustering and plotted using the Pheatmap (v1.0.12) package.

Monte-Carlo Simulation of Clonotype Restriction

All CD8 T cells were grouped according to a unique TCR clonotype. Each cell was randomly assigned either Tcirc or Trm. The number of clonotypes that were 100% restricted to either Trm or Tcirc was counted. This simulation was repeated 1e6 times. The P value was determined by the proportion of simulations that had equal or greater numbers of restricted clonotypes than what was observed in the dataset.

GLIPH2 Analysis

TCR clonotype information was extracted from the CD8 T cells. Additionally, samples were HLA types using Polysolver, Optitype, ArcasHLA, and HLA-HD from bulk and single RNA-seq data. These HLA types and the extracted clonotype information were input into the GLIPH2 online portal (http://50.255.35.37:8080) with the following parameters: GLIPH2 algorithm, reference version 2.0, CD8 reference, all_aa_interchangeable as “YES.” The results were filtered using the Fisher_score <0.05, number_unique_CDR3 >number_subject, and Freq > number_unique_CDR3.

CD8 Clonotype Database Comparison

TCR clonotype information was extracted from CD8 positive T cells: the TRB CDR3 and TRBV genes. TCRs were considered a match if both the TRB CDR3 and TRBV genes matched a TCR found in the database. The VDJdb, McPAS-TCR, and TRAdb databases were accessed on January 2, 2023 (2729).

γδ TCR Clustering

The delta CDR3 sequences were extracted from the scTCR dataset. All CDR3s were aligned pairwise to each other using the “pairwiseAlignment” function from the Biostrings package (2.62.0) with the following settings: BLOSUM62 substitution matrix, gap opening = −10, gap extension = −1. The pairwise alignments were then clustered using unsupervised hierarchical clustering.

MIXCR

We implemented the MIXCR software (ref. 36; 4.1.0) to extract T/B cell receptor sequences from the RNA-seq data. The command used was mixcr to analyze RNA-seq-tcr-full-length, with the parameters: species hsa to specify the human species and rna to indicate RNA input data. We included the -keep-non-CDR3-alignments flag to retain alignments that did not include the CDR3 region, which provides a more comprehensive perspective on the TCR repertoire. The input data consisted of paired-end RNA-seq reads. Expanded clones were defined as clones with greater than 1.5 times the reads of the least frequent clone per sample and per receptor type.

Bulk RNA-seq Processing

Sequence reads were trimmed and filtered using Trim Galore! (v0.6.7, RRID:SCR_011847) to remove low-quality reads, adapter sequences, and bases with low Phred scores. The reads were then aligned to the reference human genome (GRCh38.p13 primary assembly) augmented with the Merkel cell polyomavirus transcriptome using GENCODE v32 transcript annotation with STAR (v2.7.10b, RRID:SCR_004463) in two-pass mode. The dataset was quality controlled to remove any significant outliers. Differential gene expression analysis was performed using DESeq2 (1.28.1; RRID:SCR_000154) or other appropriate software packages. The limma package (RRID:SCR_010943) was used to fit the differential expressions to a generalized linear model. Pathway analysis was conducted with GSVA and GSEA.

Covariate DESeq

The patient metadata associated with the corresponding RNA-seq were associated with a DESeq object (RRID:SCR_000154). Raw read counts were filtered to remove low-expression genes, defined as having more than 20% of the total samples containing zero reads. A variance-stabilizing transformation was applied to the DESeq object and a PCA plot was visualized, showing clustering by virus status, tissue of origin, and patient sex. Differential expression was determined by comparing the immunotherapy response with the covariates virus status, tissue of origin, and patient sex. For pre-post ICB DE-seq, only patients were used as covariates with samples compared by time point.

Cell-Type Signature Generation

The Wilcoxon markers of the cell type of interest versus all other cells were compared to the Wilcoxon markers of that cell type compared to the next most similar cell type. Trm cells were compared to Tcirc cells and tumor cycling cells were compared to all other tumor cells. These markers were intersected and filtered for a log2 fold change >0.5 and a P value < 0.05. The genes are listed in Supplementary Table S17.

Gene Set Score

The cell-type signature genes were used as a gene set for single-sample GSVA. The corresponding GSVA scores were then classified into quartiles. The Kaplan–Meier curve for immunotherapy-specific survival compared to the top 25% of the cell-type score against the bottom 25% cell-type score among patients who had an evaluable immunotherapy response.

TCR-pMHC Complex Prediction

TCR-pMHC complex models were generated using TCRmodel2 (25) and AlphaFold (ref. 49; v.2.3.0) deep learning structural prediction methods. Structural prediction using TCRmodel2 was performed via its online server (https://tcrmodel.ibbr.umd.edu/), with structural refinement. AlphaFold 2.3.0 was downloaded from the AlphaFold Github repository (https://github.com/deepmind/alphafold) in February 2023 and run on a local computing cluster, generating 25 predictions per complex, with refinement performed on the top-ranked predictions. No template date cutoff was applied during TCRmodel2 or AlphaFold modeling. For the MO_MCC_095c1 TCR-peptide-MHC complex (Fig. 4D), models from TCRmodel2 and AlphaFold were pooled and ranked using the AlphaFold model confidence score, which is associated with TCR-pMHC model accuracy (25). The AlphaFold-generated model was ranked the highest and was selected for further inspection. For all other TCR-peptide-MHC complexes, models were generated by TCRmodel2 and ranked by model confidence score.

Data Availability

Bulk RNA-seq and scRNA-seq data are available at GEO accession GSE235093. Raw sequencing data are available at dbGAP accession phs003629 (https://www.ncbi.nlm.nih.gov/gap/sstr/report/phs003629.v1.p1).

Z.Z. Reinstein reports grants from NCI during the conduct of the study; in addition, Z.Z. Reinstein has a patent for Novel cytokines, chemokine and receptors to augment Merkel cell carcinoma immunotherapy response pending. Y. Zhang reports grants from Alpha Omega Alpha Carolyn L. Kuckein Fellowship and ISID LEO Foundation Travel Grant during the conduct of the study. S. Chandra reports other support from Bristol Myers Squibb and Regeneron during the conduct of the study; other support from Bristol Myers Squibb, Regeneron, Novartis, and Pfizer outside the submitted work. J.B. Cheng reports grants from LEO Foundation and Veterans Health Administration Office of Research and Development during the conduct of the study; grants from Sun Pharma, Regeneron, Pfizer, Bristol Myers Squibb, and Janssen outside the submitted work. C.H. Chung reports personal fees from Fulgent, GenMab, AVEO, Seagen, Regeneron, Exelixis, and Bicara during the conduct of the study. N.I. Khushalani reports grants and non-financial support from Bristol Myers-Squibb, grants, personal fees, and non-financial support from Regeneron and Replimune, grants and personal fees from Merck, personal fees from Immunocore, other support from Incyte, and AstraZeneca, personal fees and non-financial support from Iovance Biotherapeutics, grants and personal fees from Novartis, personal fees from Nektar, personal fees and non-financial support from Castle Biosciences, personal fees from Instil Bio, grants from GlaxoSmithKline, HUYA Bioscience International, Celgene, and Modulation Therapeutics, other support from Bellicum, Asensus Surgical, Amarin, and T-Knife Therapeutics outside the submitted work. A.A. Sarnaik reports personal fees from Guidepoint, Gerson Lehrman Group, second City LLC, MJH Holdings, Clinical Education Alliance, Bluprint Oncology Concepts, Boxer Capital, Rising Tide Foundation, Istari Onc, and Keyquest Health outside the submitted work; in addition, A.A. Sarnaik has a patent for 61/973,002 issued, licensed, and with royalties paid from Iovance, a patent for 61/955,970 issued, licensed, and with royalties paid from Iovance, a patent for 62/612,915 issued, a patent for 14/974,357 issued, a patent for 16/157,174 issued, and a patent for 17/279,327 issued. V.K. Sondak reports personal fees from Bristol Myers Squibb, Genesis, Iovance Biotherapeutics, Merck, Novartis, Regeneron, and Helix Biopharma, grants from Neogene Therapeutics, Skyline Dx, and Turnstone Biologics outside the submitted work. C. Vaske reports other support from NantOmics LLC during the conduct of the study. S. Kim reports grants from Bristol Myers Squibb during the conduct of the study; grants from AstraZeneca outside the submitted work. A.S. Brohl reports personal fees from Deciphera and Bayer, and grants from Merck outside the submitted work. B.L. Fridley reports grants from National Institutes of Health during the conduct of the study. K.Y. Tsai reports non-financial support from NantOmics, grants and other support from Moffitt Cancer Center Pinellas Partners and Moffitt Cancer Center Donald A. Adam Melanoma & Skin Cancer Center of Excellence, and grants from NIH during the conduct of the study; personal fees from NFlection Therapeutics, WorldCare Clinical, and Verrica Pharmaceuticals outside the submitted work; in addition, K.Y. Tsai and J. Choi has a patent for methods to augment immunotherapy response pending to Northwestern University, Moffitt Cancer Center. J. Choi reports grants from V Foundation and Leukemia and Lymphoma Society during the conduct of the study; other support from Moonlight Bio outside the submitted work; None. No disclosures were reported by the other authors.

Z.Z. Reinstein: Data curation, formal analysis, investigation, visualization, methodology, writing–original draft, writing–review and editing. Y. Zhang: Data curation, formal analysis, visualization, writing–original draft, writing–review and editing. O.E. Ospina: Data curation, formal analysis, visualization, writing–original draft, writing–review and editing. M.D. Nichols: Data curation, visualization. V.A. Chu: Investigation, visualization. A. de Mingo Pulido: Investigation. K. Prieto: Investigation. J.V. Nguyen: Formal analysis, investigation, visualization. R. Yin: Formal analysis, writing–original draft. C. Moran Segura: Formal analysis, investigation. A. Usman: Resources. B. Sell: Investigation. S. Ng: Investigation. J.V. de la Iglesia: Investigation. S. Chandra: Resources. J.A. Sosman: Resources, writing–review and editing. R.J. Cho: Methodology. J.B. Cheng: Methodology. E. Ivanova: Methodology. S.B. Koralov: Methodology. R.J. C. Slebos: Investigation. C.H. Chung: Investigation. N.I. Khushalani: Resources. J.L. Messina: Resources. A.A. Sarnaik: Resources. J.S. Zager: Resources. V.K. Sondak: Resources. C. Vaske: Investigation. S. Kim: Resources. A.S. Brohl: Resources. X. Mi: Formal analysis. B.G. Pierce: Formal analysis, supervision. X. Wang: Formal analysis. B.L. Fridley: Formal analysis, supervision. K.Y. Tsai: Conceptualization, resources, formal analysis, supervision, funding acquisition, writing–original draft, project administration, writing–review and editing. J. Choi: Conceptualization, resources, formal analysis, supervision, funding acquisition, writing–original draft, project administration, writing–review and editing.

This research was made possible through the Division of Research Pathology, Total Cancer Care protocol and supported by the Advanced Analytical Digital Laboratory, Tissue Core, Flow Cytometry Core, and Molecular Genomics Core at the H. Lee Moffitt Cancer Center & Research Institute, an NCI-designated Comprehensive Cancer Center (P30-CA076292). Funding support was provided by the Donald A. Adam Melanoma & Skin Cancer Center of Excellence (K.Y. Tsai), the Barry S. Greene Fund (K.Y. Tsai, A.S. Brohl), and the Moffitt Distinguished Scholar Award (N.I. Khushalani). The authors acknowledge the Nanostring Technology Access Program for access to both the GeoMx and CosMx platforms to generate spatial transcriptomic data. We would like to thank the Northwestern Dermatology Clinical Trials Unit for their assistance with tissue acquisition. This work was supported by the Northwestern University Flow Cytometry Core Facility and the Cancer Center Support Grant (NCI CA060553). The research reported in this publication was supported by the Northwestern University Skin Biology & Diseases Resource-Based Center of the National Institutes of Health under the award number P30AR075049. Funding for this study was provided by the National Cancer Institute grant T32 CA009560 and F30CA278298 (Z.Z. Reinstein), National Cancer Institute grant T32 CA233399 (O.E. Ospina, B.L. Fridley), the 2019 AOA Carolyn E. Kuckein Fellowship (Y. Zhang), the V Foundation for Cancer Research grant T2021-019 (J. Choi), the Bakewell Foundation (J. Choi), National Institutes of Health grants DP2 OD024475-01 (J. Choi), the National Comprehensive Cancer Network YIA (J. Choi), R35 GM144083 (B. Pierce), and the Leukemia and Lymphoma Society grant 1377-21 (J. Choi).

Note Supplementary data for this article are available at Cancer Discovery Online (http://cancerdiscovery.aacrjournals.org/).

1.
McEvoy
AM
,
Lachance
K
,
Hippe
DS
,
Cahill
K
,
Moshiri
Y
,
Lewis
CW
, et al
.
Recurrence and mortality risk of Merkel cell carcinoma by cancer stage and time from diagnosis
.
JAMA Dermatol
2022
;
158
:
382
9
.
2.
Knepper
TC
,
Montesion
M
,
Russell
JS
,
Sokol
ES
,
Frampton
GM
,
Miller
VA
, et al
.
The genomic landscape of Merkel cell carcinoma and clinicogenomic biomarkers of response to immune checkpoint inhibitor therapy
.
Clin Cancer Res
2019
;
25
:
5961
71
.
3.
DeCaprio
JA
.
Molecular pathogenesis of Merkel cell carcinoma
.
Annu Rev Pathol
2021
;
16
:
69
91
.
4.
Goh
G
,
Walradt
T
,
Markarov
V
,
Blom
A
,
Riaz
N
,
Doumani
R
, et al
.
Mutational landscape of MCPyV-positive and MCPyV-negative Merkel cell carcinomas with implications for immunotherapy
.
Oncotarget
2016
;
7
:
3403
15
.
5.
Lahman
MC
,
Paulson
KG
,
Nghiem
PT
,
Chapuis
AG
.
Quality is king: fundamental insights into tumor antigenicity from virus-associated Merkel cell carcinoma
.
J Invest Dermatol
2021
;
141
:
1897
905
.
6.
Nghiem
P
,
Bhatia
S
,
Lipson
EJ
,
Sharfman
WH
,
Kudchadkar
RR
,
Brohl
AS
, et al
.
Three-year survival, correlates and salvage therapies in patients receiving first-line pembrolizumab for advanced Merkel cell carcinoma
.
J Immunother Cancer
2021
;
9
:
e002478
.
7.
D’Angelo
SP
,
Lebbé
C
,
Mortier
L
,
Brohl
AS
,
Fazio
N
,
Grob
J-J
, et al
.
First-line avelumab in a cohort of 116 patients with metastatic Merkel cell carcinoma (JAVELIN Merkel 200): primary and biomarker analyses of a phase II study
.
J Immunother Cancer
2021
;
9
:
e002646
.
8.
Paulson
KG
,
Voillet
V
,
McAfee
MS
,
Hunter
DS
,
Wagener
FD
,
Perdicchio
M
, et al
.
Acquired cancer resistance to combination immunotherapy from transcriptional loss of class I HLA
.
Nat Commun
2018
;
9
:
3868
.
9.
Chen
EY
,
Tan
CM
,
Kou
Y
,
Duan
Q
,
Wang
Z
,
Meirelles
GV
, et al
.
Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool
.
BMC Bioinformatics
2013
;
14
:
128
.
10.
Grasso
CS
,
Tsoi
J
,
Onyshchenko
M
,
Abril-Rodriguez
G
,
Ross-Macdonald
P
,
Wind-Rotolo
M
, et al
.
Conserved interferon-γ signaling drives clinical response to immune checkpoint blockade therapy in melanoma
.
Cancer Cell
2020
;
38
:
500
15.e3
.
11.
Huang
R
,
Grishagin
I
,
Wang
Y
,
Zhao
T
,
Greene
J
,
Obenauer
JC
, et al
.
The NCATS BioPlanet—an integrated platform for exploring the universe of cellular signaling pathways for toxicology, systems biology, and chemical genomics
.
Front Pharmacol
2019
;
10
:
445
.
12.
Tirosh
I
,
Izar
B
,
Prakadan
SM
,
Wadsworth
MH
II
,
Treacy
D
,
Trombetta
JJ
, et al
.
Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq
.
Science
2016
;
352
:
189
96
.
13.
Reizis
B
.
Plasmacytoid dendritic cells: development, regulation, and function
.
Immunity
2019
;
50
:
37
50
.
14.
Hegde
PS
,
Chen
DS
.
Top 10 challenges in cancer immunotherapy
.
Immunity
2020
;
52
:
17
35
.
15.
Van de Sande
B
,
Flerin
C
,
Davie
K
,
De Waegeneer
M
,
Hulselmans
G
,
Aibar
S
, et al
.
A scalable SCENIC workflow for single-cell gene regulatory network analysis
.
Nat Protoc
2020
;
15
:
2247
76
.
16.
Coughlan
E
,
Garside
VC
,
Wong
SFL
,
Liang
H
,
Kraus
D
,
Karmakar
K
, et al
.
A hox code defines spinocerebellar neuron subtype regionalization
.
Cell Rep
2019
;
29
:
2408
21.e4
.
17.
Nefzger
CM
,
Haynes
JM
,
Pouton
CW
.
Directed expression of Gata2, Mash1, and Foxa2 synergize to induce the serotonergic neuron phenotype during in vitro differentiation of Embryonic stem cells
.
Stem Cells
2011
;
29
:
928
39
.
18.
Bunt
J
,
Hasselt
NE
,
Zwijnenburg
DA
,
Hamdi
M
,
Koster
J
,
Versteeg
R
, et al
.
OTX2 directly activates cell cycle genes and inhibits differentiation in medulloblastoma cells
.
Int J Cancer
2012
;
131
:
E21
32
.
19.
van der Leun
AM
,
Thommen
DS
,
Schumacher
TN
.
CD8+ T cell states in human cancer: insights from single-cell analysis
.
Nat Rev Cancer
2020
;
20
:
218
32
.
20.
Yenyuwadee
S
,
Sanchez-Trincado Lopez
JL
,
Shah
R
,
Rosato
PC
,
Boussiotis
VA
.
The evolving role of tissue-resident memory T cells in infections and cancer
.
Sci Adv
2022
;
8
:
eabo5871
.
21.
Tokura
Y
,
Phadungsaksawasdi
P
,
Kurihara
K
,
Fujiyama
T
,
Honda
T
.
Pathophysiology of skin resident memory T cells
.
Front Immunol
2021
;
11
:
618897
.
22.
Heczey
A
,
Xu
X
,
Courtney
AN
,
Tian
G
,
Barragan
GA
,
Guo
L
, et al
.
Anti-GD2 CAR-NKT cells in relapsed or refractory neuroblastoma: updated phase 1 trial interim results
.
Nat Med
2023
;
29
:
1379
88
.
23.
Lowery
FJ
,
Krishna
S
,
Yossef
R
,
Parikh
NB
,
Chatani
PD
,
Zacharakis
N
, et al
.
Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers
.
Science
2022
;
375
:
877
84
.
24.
Iyer
JG
,
Afanasiev
OK
,
McClurkan
C
,
Paulson
K
,
Nagase
K
,
Jing
L
, et al
.
Merkel cell polyomavirus-specific CD8+ and CD4+ T-cell responses identified in Merkel cell carcinomas and blood
.
Clin Cancer Res
2011
;
17
:
6671
80
.
25.
Yin
R
,
Ribeiro-Filho
HV
,
Lin
V
,
Gowthaman
R
,
Cheung
M
,
Pierce
BG
.
TCRmodel2: high-resolution modeling of T cell receptor recognition using deep learning
.
Nucleic Acids Res
2023
;
51
:
W569
76
.
26.
Huang
H
,
Wang
C
,
Rubelt
F
,
Scriba
TJ
,
Davis
MM
.
Analyzing the Mycobacterium tuberculosis immune response by T-cell receptor clustering with GLIPH2 and genome-wide antigen screening
.
Nat Biotechnol
2020
;
38
:
1194
202
.
27.
Goncharov
M
,
Bagaev
D
,
Shcherbinin
D
,
Zvyagin
I
,
Bolotin
D
,
Thomas
PG
, et al
.
VDJdb in the pandemic era: a compendium of T cell receptors specific for SARS-CoV-2
.
Nat Methods
2022
;
19
:
1017
9
.
28.
Tickotsky
N
,
Sagiv
T
,
Prilusky
J
,
Shifrut
E
,
Friedman
N
.
McPAS-TCR: a manually curated catalogue of pathology-associated T cell receptor sequences
.
Bioinformatics
2017
;
33
:
2924
9
.
29.
Zhang
W
,
Wang
L
,
Liu
K
,
Wei
X
,
Yang
K
,
Du
W
, et al
.
PIRD: pan immune repertoire database
.
Bioinformatics
2020
;
36
:
897
903
.
30.
Mimitou
EP
,
Cheng
A
,
Montalbano
A
,
Hao
S
,
Stoeckius
M
,
Legut
M
, et al
.
Multiplexed detection of proteins, transcriptomes, clonotypes and CRISPR perturbations in single cells
.
Nat Methods
2019
;
16
:
409
12
.
31.
Daniels
J
,
Doukas
PG
,
Escala
MEM
,
Ringbloom
KG
,
Shih
DJH
,
Yang
J
, et al
.
Cellular origins and genetic landscape of cutaneous gamma delta T cell lymphomas
.
Nat Commun
2020
;
11
:
1806
.
32.
Gherardin
NA
,
Waldeck
K
,
Caneborg
A
,
Martelotto
LG
,
Balachander
S
,
Zethoven
M
, et al
.
γδ T cells in Merkel cell carcinomas have a proinflammatory profile prognostic of patient survival
.
Cancer Immunol Res
2021
;
9
:
612
23
.
33.
Davey
MS
,
Willcox
CR
,
Hunter
S
,
Kasatskaya
SA
,
Remmerswaal
EBM
,
Salim
M
, et al
.
The human Vδ2+ T-cell compartment comprises distinct innate-like Vγ9+ and adaptive Vγ9 subsets
.
Nat Commun
2018
;
9
:
1760
.
34.
Creed
JH
,
Wilson
CM
,
Soupir
AC
,
Colin-Leitzinger
CM
,
Kimmel
GJ
,
Ospina
OE
, et al
.
spatialTIME and iTIME: R package and Shiny application for visualization and analysis of immunofluorescence data
.
Bioinformatics
2021
;
37
:
4584
6
.
35.
Jin
S
,
Guerrero-Juarez
CF
,
Zhang
L
,
Chang
I
,
Ramos
R
,
Kuan
C-H
, et al
.
Inference and analysis of cell-cell communication using CellChat
.
Nat Commun
2021
;
12
:
1088
.
36.
Bolotin
DA
,
Poslavsky
S
,
Mitrophanov
I
,
Shugay
M
,
Mamedov
IZ
,
Putintseva
EV
, et al
.
MiXCR: software for comprehensive adaptive immunity profiling
.
Nat Methods
2015
;
12
:
380
1
.
37.
Li
H
,
van der Leun
AM
,
Yofe
I
,
Lubling
Y
,
Gelbard-Solodkin
D
,
van Akkooi
ACJ
, et al
.
Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma
.
Cell
2019
;
176
:
775
89.e18
.
38.
Pulliam
T
,
Jani
S
,
Jing
L
,
Ryu
H
,
Jojic
A
,
Shasha
C
, et al
.
Circulating cancer-specific CD8 T cell frequency is associated with response to PD-1 blockade in Merkel cell carcinoma
.
Cell Rep Med
2024
;
5
:
101412
.
39.
Weeden
CE
,
Gayevskiy
V
,
Marceaux
C
,
Batey
D
,
Tan
T
,
Yokote
K
, et al
.
Early immune pressure initiated by tissue-resident memory T cells sculpts tumor evolution in non-small cell lung cancer
.
Cancer Cell
2023
;
41
:
837
52.e6
.
40.
de Vries
NL
,
van de Haar
J
,
Veninga
V
,
Chalabi
M
,
Ijsselsteijn
ME
,
van de Ploeg
M
, et al
.
γδ T cells are effectors of immunotherapy in cancers with HLA class I defects
.
Nature
2023
;
613
:
743
50
.
41.
Wu
Y
,
Kyle-Cezar
F
,
Woolf
RT
,
Naceur-Lombardelli
C
,
Owen
J
,
Biswas
D
, et al
.
An innate-like Vδ1+ γδ T cell compartment in the human breast is associated with remission in triple-negative breast cancer
.
Sci Transl Med
2019
;
11
:
eaax9364
.
42.
Oshi
M
,
Newman
S
,
Tokumaru
Y
,
Yan
L
,
Matsuyama
R
,
Kalinski
P
, et al
.
Plasmacytoid dendritic cell (pDC) infiltration correlate with tumor infiltrating lymphocytes, cancer immunity, and better survival in triple negative breast cancer (TNBC) more strongly than conventional dendritic cell (cDC)
.
Cancers (Basel)
2020
;
12
:
3342
.
43.
Conrad
C
,
Gregorio
J
,
Wang
Y-H
,
Ito
T
,
Meller
S
,
Hanabuchi
S
, et al
.
Plasmacytoid dendritic cells promote immunosuppression in ovarian cancer via ICOS costimulation of Foxp3+ T-regulatory cells
.
Cancer Res
2012
;
72
:
5240
9
.
44.
Boersma
B
,
Jiskoot
W
,
Lowe
P
,
Bourquin
C
.
The interleukin-1 cytokine family members: role in cancer pathogenesis and potential therapeutic applications in cancer immunotherapy
.
Cytokine Growth Factor Rev
2021
;
62
:
1
14
.
45.
He
S
,
Bhatt
R
,
Brown
C
,
Brown
EA
,
Buhr
DL
,
Chantranuvatana
K
, et al
.
High-plex imaging of RNA and proteins at subcellular resolution in fixed tissue by spatial molecular imaging
.
Nat Biotechnol
2022
;
40
:
1794
806
.
46.
Mann
SE
,
Zhou
Z
,
Landry
LG
,
Anderson
AM
,
Alkanani
AK
,
Fischer
J
, et al
.
Multiplex T cell stimulation assay utilizing a T cell activation reporter-based detection System
.
Front Immunol
2020
;
11
:
633
.
47.
Heemskerk
MHM
,
Hoogeboom
M
,
de Paus
RA
,
Kester
MGD
,
van der Hoorn
MAWG
,
Goulmy
E
, et al
.
Redirection of antileukemic reactivity of peripheral T lymphocytes using gene transfer of minor histocompatibility antigen HA-2-specific T-cell receptor complexes expressing a conserved alpha joining region
.
Blood
2003
;
102
:
3530
40
.
48.
Fleming
SJ
,
Chaffin
MD
,
Arduini
A
,
Akkad
A-D
,
Banks
E
,
Marioni
JC
, et al
.
Unsupervised removal of systematic background noise from droplet-based single-cell experiments using CellBender
.
Nat Methods
2023
;
20
:
1323
35
.
49.
Jumper
J
,
Evans
R
,
Pritzel
A
,
Green
T
,
Figurnov
M
,
Ronneberger
O
, et al
.
Highly accurate protein structure prediction with AlphaFold
.
Nature
2021
;
596
:
583
9
.

Supplementary data